US20070249621A1 - Pharmacological treatment of parkinson's disease - Google Patents

Pharmacological treatment of parkinson's disease Download PDF

Info

Publication number
US20070249621A1
US20070249621A1 US11/713,156 US71315607A US2007249621A1 US 20070249621 A1 US20070249621 A1 US 20070249621A1 US 71315607 A US71315607 A US 71315607A US 2007249621 A1 US2007249621 A1 US 2007249621A1
Authority
US
United States
Prior art keywords
receptor agonist
agonist
dopamine
ht1a
dopamine receptor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/713,156
Other versions
US9066903B2 (en
Inventor
William Wolf
Erik Istre
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Veterans Affairs VA
Original Assignee
US Department of Veterans Affairs VA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by US Department of Veterans Affairs VA filed Critical US Department of Veterans Affairs VA
Priority to US11/713,156 priority Critical patent/US9066903B2/en
Assigned to UNITED STATES GOVERNMENT AS REPRESENTED BY THE DEPARTMENT OF VETERANS AFFAIRS reassignment UNITED STATES GOVERNMENT AS REPRESENTED BY THE DEPARTMENT OF VETERANS AFFAIRS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WOLF, WILLIAM A., ISTRE, ERIK D.
Publication of US20070249621A1 publication Critical patent/US20070249621A1/en
Priority to US12/730,972 priority patent/US9226904B2/en
Application granted granted Critical
Publication of US9066903B2 publication Critical patent/US9066903B2/en
Priority to US14/958,430 priority patent/US20160082006A1/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs

Definitions

  • the present invention relates to the co-administration of two neurotransmitter agonists to patients with motor disorders, for the purpose of symptom reduction.
  • the present invention provides methods and compositions for alleviation of akinesia, rigidity and/or tremor associated with Parkinson's disease.
  • Parkinson's disease is a chronic, progressive, hypokinetic disorder characterized by impaired voluntary movement (See, Dale and Federman (eds.), WebMD Scientific American Medicine , NY: WebMD Corporation, Chapter 11, Section 15, pp. 1-21, 2001; Lang and Lozano, N Engl J Med, 339:1044, 1998; and Lang and Lozano, N Engl J Med, 339:1130, 1998). Parkinson's disease occurs as a result of the death of dopamine-producing neurons in the substantia nigra of the midbrain. Dopamine is a neurotransmitter, or chemical messenger, that transports signals to the parts of the brain that control movement initiation and coordination.
  • the loss of dopamine in the brain is associated with multiple primary symptoms including: tremor of the hands, arms, legs, jaw, and face; rigidity or stiffness of the limbs and trunk; bradykinesia or slowness of movement; and postural instability or impaired balance and coordination.
  • Parkinson's disease afflicts more than one million persons in the United States alone (Lang and Lozano, supra, 1998), with approximately 50,000 new cases diagnosed each year. It is generally a disease of late middle age, with typical onset occurring at about age 60. About five percent of patients, however, have early-onset disease and are younger than 40 when symptoms begin.
  • L-DOPA dopamine precursor
  • PD Parkinson's disease
  • the dopamine precursor, levodopa (L-DOPA) is still considered to be the gold standard in terms of treatment for PD (Schapira and Olanow, JAMA, 291:358-364, 2004).
  • L-DOPA can cause debilitating side effects (LeWitt and Nyholm Neurology, 62:S9-S16, 2004), including severe nausea, vomiting, and psychosis.
  • patients frequently experience other side effects such as dyskinesias (spontaneous, involuntary movements) and “on-off” periods when the medication will suddenly start or stop working.
  • Surgical treatments are considered for patients with advanced disease or who have not responded adequately to medications. Accepted surgical treatments involve either the creation of small, precise lesions or the implantation of stimulating electrodes in specific brain regions that appear to be overactive in Parkinson's disease. More recently, implantation of fetal dopaminergic tissue has been utilized as a means of restoring dopamine levels in the brains of patients with Parkinson's disease (See, e.g., Freed et al., Arch Neurol, 47:505-512, 1990; and Lindvall et al., Science, 247:574-577, 1990). These types of methods, however, are highly invasive.
  • the present invention relates to the co-administration of two neurotransmitter agonists to patients with motor disorders, for the purpose of symptom reduction.
  • the present invention provides methods and compositions for alleviation of akinesia, rigidity and/or tremor associated with Parkinson's disease, as well as for alleviation of side effects associated with treatment associated with Parkinson's disease.
  • the present invention provides methods of treating a motor disorder, comprising: providing: i) a serotonin type 1A (5-HT1A) receptor agonist, ii) a dopamine receptor agonist, and iii) a subject diagnosed as having a motor disorder; and b) administering the serotonin type 1A receptor (5-HT1A) agonist to the subject during a pre-conditioning period; and c) co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with a low dose of the dopamine receptor agonist to the subject, under conditions suitable for reducing at least one symptom of the motor disorder, wherein the low dose of the dopamine receptor agonist is below that required for reducing at least one symptom of the motor disorder when administered in the absence of the serotonin type 1A (5-HT1A) receptor agonist.
  • the motor disorder is Parkinson's disease. In other embodiments, the motor disorder is parkinsonism arising from viral infection or drug use.
  • the dopamine receptor agonist is not a dopamine precursor.
  • the serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of arylpiperazines, azapirones, aminotetralins, and aminochromanes. In other embodiments, the serotonin type 1A (5-HT1A) receptor agonist is 8-hydroxy-2-(di-n-propylamino)tetralin (8OH-DPAT). In some preferred embodiments, 8OH-DPAT is administered via a transdermal patch.
  • the serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of tandospirone, ipsapirone, gepirone, flibanserin, flesinoxan, sarizotan, repinotan and PRX-00023.
  • the dopamine receptor agonist is a D1 type dopamine receptor agonist.
  • the D1 type dopamine receptor agonist is selected from the group consisting of SKF38393, SKF83959, SKF81297, SKF77434, SKF75670, SKF82958, and derivatives thereof.
  • the D1 type dopamine receptor agonist is selected from the group consisting of dihydrexidine, dinapsoline, A-77636, ABT-431, CY208-243, and A-68930.
  • the dopamine receptor agonist is a D2 type dopamine receptor agonist.
  • the D2 type dopamine receptor agonist is quinpirole.
  • the D2 type dopamine receptor agonist is selected from the group consisting of quinelorane, terguride, pergolide, talipexole, pramipexole, sumanirole, ropinirole, roxindole, bromocriptine, and cabergoline.
  • the dopamine agonist is a dopamine precursor.
  • the dopamine precursor is levodopa (L-DOPA), which is administered with one or both of a L-aromatic amino acid decarboxylase inhibitor (e.g., carbidopa, benserazide, difluromethyldopa, a-methyldopa, etc.) and a catechol-O-methyltransferase (COMT) inhibitor (e.g., tolcapone and entacapone).
  • L-DOPA levodopa
  • L-DOPA L-aromatic amino acid decarboxylase inhibitor
  • COT catechol-O-methyltransferase
  • one or both of the serotonin type 1A (5-HT1A) receptor agonist and the dopamine receptor agonist is a D1 type dopamine receptor agonist.
  • the serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of sunipetron and aripiprazole.
  • the present invention provides methods of treating Parkinson's disease, comprising: providing: i) a serotonin type 1A (5-HT1A) receptor agonist, ii) a dopamine receptor agonist, and iii) a subject diagnosed as having Parkinson's disease; and b) administering the serotonin type 1A receptor (5-HT1A) agonist to the subject during a pre-conditioning period; and c) co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with a low dose of the dopamine receptor agonist to the subject, under conditions suitable for reducing at least one symptom of Parkinson's disease, wherein the low dose of the dopamine receptor agonist is below that required for reducing at least one symptom of Parkinson's disease when administered in the absence of the serotonin type 1A (5-HT1A) receptor agonist.
  • the at least one symptom comprises one or more of bradykinesia, tremor, and muscle rigidity.
  • the co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with the dopamine receptor agonist to the subject is under conditions further suitable for reducing at least one side effect of the dopamine receptor agonist.
  • the at least one side effect of the dopamine receptor agonist comprises dyskinesia (e.g., one or more of chewing, gnawing, twisting, tongue or mouth movements, head bobbing, and movements of the feet, hands, or shoulder).
  • the at least one side effect of the dopamine receptor agonist comprises a psychiatric disturbance (e.g., one or more of memory loss, anxiety, nervousness, agitation, restlessness, confusion, inability to sleep, nightmares, daytime tiredness, mental depression and euphoria).
  • a psychiatric disturbance e.g., one or more of memory loss, anxiety, nervousness, agitation, restlessness, confusion, inability to sleep, nightmares, daytime tiredness, mental depression and euphoria.
  • the dopamine receptor agonist is not a dopamine precursor.
  • the present invention provides methods for treating a subject, comprising: providing; i) a serotonin type 1A (5-HT1A) receptor agonist, ii) a dopamine receptor agonist, iii) a subject receiving the dopamine receptor agonist for treatment of Parkinson's disease, wherein the subject is suffering from dopamine receptor agonist-induced dyskinesia; co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with the dopamine receptor agonist to the subject during a preconditioning period under conditions suitable for reducing the dopamine receptor agonist-induced dyskinesia; co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with the dopamine receptor agonist to the subject during a dosage optimization period, wherein dosage of the dopamine receptor agonist is gradually reduced until at least one symptom of the Parkinson's disease is re-emerges; and co-administering the serotonin type 1A
  • kits for treating Parkinson's disease comprising a serotonin type 1A ( 5 -HT1A) receptor agonist, a dopamine receptor agonist, and instructions for administering the serotonin type 1A ( 5 -HT1A) receptor agonist and the dopamine receptor agonist to a subject diagnosed as having Parkinson's disease, wherein the dopamine receptor agonist is administered at a dose below that required for reducing at least one symptom of the Parkinson's disease when administered in the absence of the serotonin type 1A (5-HT1A) receptor agonist.
  • the kit comprises a package (e.g., blister pack).
  • the dopamine receptor agonist is not a dopamine precursor.
  • compositions for treating Parkinson's disease comprising: a therapeutic dose of a serotonin type 1A (5-HT1A) receptor agonist and a subtherapeutic dose of a dopamine receptor agonist.
  • the dopamine receptor agonist is not a dopamine precursor.
  • the compositions further comprise a pharmaceutically acceptable excipient.
  • FIG. 1 depicts the effects of repeated daily dosing with L-DOPA (10 mg/kg+6.25 mg/kg benserazide) or L-DOPA plus R(+)-8-OHDPAT (0.3 mg/kg). Ipsilateral turning as well as contralateral turning was recorded and quantified.
  • FIG. 2 demonstrates that a 5-HT 1A agonist enhances the anti-parkinsonian action of a D 1 type dopamine agonist as demonstrated by contralateral turning in the unilateral 6-OHDA-lesioned rat.
  • the 5-HT 1A agonist R(+)-8-OHDPAT 0.3 mg/kg or vehicle (0.9% saline containing ascorbate) was administered for 14 days.
  • rats were given either vehicle or R(+)-8-OHDPAT (0.3 mg/kg), plus a low dose D 1 agonist SKF 38393 (0.1 mg/kg) and observed for 3 hr.
  • FIG. 3 demonstrates that a 5-HT 1A agonist enhances the anti-parkinsonian action of a D 2 type dopamine agonist as demonstrated by contralateral turning in the unilateral 6-OHDA-lesioned rat.
  • the 5-HT 1A agonist R(+)-8-OHDPAT (0.3 mg/kg) or vehicle (0.9% saline containing ascorbate) was administered for 14 days.
  • rats were given either vehicle or R(+)-8-OHDPAT (0.3 mg/kg), plus a low dose D 2 agonist quinpirole (0.05 mg/kg) and observed for 3 hr.
  • FIG. 4 illustrates that both repeated and concomitant administration of a serotonin agonist was required to enhance that anti-parkinsonian actions of a dopamine agonist.
  • FIG. 5 demonstrates that repeated administration of R(+)-8-OHDPAT reduced dyskinetic behavior elicited by a moderately high dose of the D 1 agonist SKF 38393 (3.0 mg/kg).
  • FIG. 6 demonstrates that repeated administration of R(+)-8-OHDPAT reduced dyskinetic behavior elicited by a moderately high dose of the D 2 agonist quinpirole (0.05 mg/kg).
  • FIG. 7 illustrates that pre-treatment with a selective 5-HT1A antagonist (WAY 100635) blocked the facilitating effects of a serotonin agonist on the anti-parkinsonian action of a dopamine agonist.
  • FIG. 8 is an image of an immunoblot of striatal extracts probed with an anti-Fos antibody.
  • FIG. 9 illustrates that once daily administration of L-DOPA (25 mg/kg plus 6.25 mg/kg benserazide) or SKF 38393 (1 mg/kg) for 15 days elicits a significant increase in ⁇ FosB in the denervated striatum of unilateral 6-OHDA-lesioned rats (*, significantly greater than vehicle-treated; p ⁇ 0.05).
  • the concomitant administration of the 5-HT1A agonist R(+)-8-OHDPAT (0.3 mg/kg) with SKF 38393 prevents the D1 agonist-induced rise in ⁇ FosB (#, significantly less than VEH+SKF; p ⁇ 0.05 and not significantly different from vehicle-treated).
  • FIG. 10 depicts the rotational response in rats given once daily administration of vehicle+SKF 38393 (0.1, 0.3 or 1.0 mg/kg) for 15 days.
  • Animals receiving once daily R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 displayed a significantly greater contralateral turning response that displayed a dose-dependency with respect to the dose of SKF administered.
  • FIG. 11 compares the rotational response between pre-screened (“primed”) and non-pre-screened (“unprimed”) rats after 15 days of once daily administration of R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 (0.3 mg/kg). There is no significant difference between the groups.
  • Parkinson's disease refers to a neurological syndrome characterized by a dopamine deficiency, resulting from degenerative, vascular, or inflammatory changes in the basal ganglia of the substantia nigra. This term also refers to a syndrome which resembles Parkinson's disease, but which may or may not be caused by Parkinson's disease, such as Parkinsonian-like side effects caused by certain antipsychotic drugs. Parkinson's disease is also referred to as paralysis agitans and shaking palsy.
  • the term “early stage of Parkinson's disease” refers broadly to the first stages in Parkinson's disease, wherein a person suffering from the disease exhibits mild symptoms that are not disabling, such as an episodic tremor of a single limb (e.g., the hand), and which affect only one side of the body.
  • advanced stage of Parkinson's disease refers broadly to a more progressive stage in Parkinson's disease, wherein a person suffering from the disease exhibits symptoms which are typically severe and which may lead to some disability (e.g., tremors encompassing both sides of the body, balance problems, etc.). Symptoms associated with advanced stage Parkinson's disease may vary significantly in individuals, and may take many years to manifest after the initial appearance of the disease.
  • subject suffering from a Parkinson's disease and “subject suffering from parkinsonism” as used herein, refer to both humans and animals displaying one or more symptom of Parkinson's disease.
  • the primary symptoms of Parkinson's disease include bradykinesia, tremor, and muscle rigidity.
  • the term “animals” refers to all non-human animals. Such non-human animals include, but are not limited to, vertebrates such as rodents, non-human primates, ovines, bovines, lagomorphs, porcines, caprines, equines, canines, felines, aves, etc.
  • dopamine refers to a catecholamine neurotransmitter and hormone, formed first by the hydroxylation of the amino acid tyrosine to dihydroxyphenylalanine (dopa), and then by decarboxylation of dihydroxyphenylalanine (dopa) to dopamine.
  • Dopamine is a precursor of adrenaline (epinephrine) and noradrenaline (norepinephrine).
  • Serotonin refers to a phenolic amine neurotransmitter produced from tryptophan by hydroxylation and decarboxylation in serotonergic neurons of the central nervous system and enterochromaffin cells of the gastrointestinal tract. Serotonin is a precursor of melatonin.
  • symptoms refers to the outward manifestations of a disease.
  • patients with Parkinson's disease exhibit symptoms such as bradykinesia, tremor, and muscle rigidity. Additional symptoms but are not limited to atypical gait, postural instability, and loss of balance.
  • the term “alleviating” refers to the act of providing relief from some painful state.
  • the term “alleviating” refers to the lessoning of symptoms of Parkinson's disease or to the lessoning of side effects associated with long term L-dopa administration.
  • the term “alleviating” comprises curing Parkinson's disease.
  • the term “alleviating” comprises decreasing the severity of the symptoms of Parkinson's disease as measured by any one of several accepted diagnostic measures including but not limited to UPDRS, Schwab and England Scale, and CAPIT.
  • Unified Parkinson's Disease Rating Scale and “UPDRS” refer to a standardized tool used to measure Parkinson's Disease severity, as described by Fahn et al., in Recent Developments in Parkinson's Disease , Fahn et al. (eds.) Plurham Park, N.J.: Macmillian Healthcare Information, 2:153-163, 1987.
  • “Schwab and England Scale” refers to a standardized tool used to measure the efficacy of surgical treatment for Parkinson's Disease, as described by Schwab and England, in Third Symposium on Parkinson's Disease , Gillingham and Donaldon (eds.) Edinburgh, Scotland: Livingstone, pp. 152-157, 1969.
  • Core Assessment Program for Intracerebral Transplantation and “CAPIT” refer to a standardized tool used to measure the efficacy of cerebral surgery, as described by Langston et al., Mov Disord, 7:2-13, 1992.
  • agonist and “stimulator” refers to molecules or compounds that mimic the action of a “native” or “natural” compound. Agonists may be homologous to these natural compounds in respect to conformation, charge or other characteristics. Thus, agonists may be recognized by receptors expressed on cell surfaces. This recognition may result in physiologic and/or biochemical changes within the cell, such that the cell reacts to the presence of the agonist in the same manner as if the natural compound was present.
  • agonist also encompasses the class of agents known as partial agonists, in which the maximal physiologic and/or biochemical changes that occur upon partial agonist administration may be less than the maximal effect exhibited by full agonists or the natural compound.
  • the term agonist refers to molecules that bind to and activate one or both of serotonin type 1A and dopamine type receptors.
  • antagonists refer to molecules or compounds that inhibit the action of a “native” or “natural” compound. Antagonists may or may not be homologous to these natural compounds in respect to conformation, charge or other characteristics. Thus, antagonists may be recognized by the same or different receptors than are recognized by an agonist. Antagonists may have allosteric effects, which prevent the action of an agonist. In contrast to the agonists, antagonistic compounds do not result in physiologic and/or biochemical changes within the cell, such that the cell reacts to the presence of the antagonist in the same manner as if the natural compound was absent.
  • a subtherapeutic dose refers to a quantity of a neurotransmitter agonist (e.g., D1 agonist) below that used to treat disease when used as a monotherapy.
  • a subtherapeutic dose of a D1 agonist is equivalent to 5 mg of SKF38393 administered to an adult human subject four times a day.
  • a therapeutic dose refers to a quantity of a neurotransmitter agonist (e.g., 5-HT1A agonist) used to treat disease (e.g., effect the cure of or that will correct the manifestations of a disease).
  • a therapeutic dose of a 5-HT1A agonist is equivalent to 5 mg of sarizotan administered to an adult human subject two or three times per day.
  • a therapeutic dose of a 5-HT1A agonist is equivalent to 5 mg of ipsapirone administered to an adult human subject four times a day.
  • the present invention relates to the co-administration of two neurotransmitter agonists to patients with motor disorders, for the purpose of symptom reduction.
  • the present invention provides methods and compositions for alleviation of akinesia, rigidity and/or tremor associated with Parkinson's disease.
  • Parkinson's disease (PD) and related neurological disorders involve the progressive degeneration of dopamine neurons innervating the striatum.
  • the loss of dopamine leads to excessive inhibition of thalamocortical motor activity, for which the primary manifestations are rigidity, bradykinesia, tremor and postural instability (Fahn, Ann NY Acad Sci, 991:1-14, 2003; and Hamani and Lozano, Ann NY Acad Sci, 991:15-21, 2003).
  • the striatum is a major input structure that receives dense dopaminergic projections from the substantia nigra pars compacta and glutamatergic projections from the cortex and thalamus.
  • Two efferent pathways originate in the striatum and utilize the inhibitory transmitter gamma-aminobutyric acid (GABA).
  • GABA inhibitory transmitter gamma-aminobutyric acid
  • the direct pathway is comprised of medium spiny striatonigral neurons that project directly to the output nuclei of the basal ganglia, which are the internal globus pallidus (rat homologue is the entopeduncular nucleus or EP) and the substantia nigra pars reticulata (SNr).
  • striatonigral neurons predominantly express dopamine D 1 receptors and co-express dynorphin and substance P (Gerfen, Clin Neuropharmacol, 18:S162-S177, 1995).
  • the indirect pathway is comprised of medium spiny striatopallidal neurons that project to the external globus pallidus (GP).
  • GP globus pallidus
  • These striatopallidal neurons predominantly express dopamine D 2 receptors and co-express enkephalin (Gerfen, supra, 1995).
  • the GP sends GABA projections to the subthalamic nucleus (STN), which in turn, sends glutamate projections to the EP and SNr.
  • STN subthalamic nucleus
  • the EP and SNr serve as the final output structures of the basal ganglia and innervate the motor thalamus with inhibitory GABA projections.
  • PD and pre-clinical models of PD there is evidence of de-synchronization and hyperactivity in the STN, EP and SNr (Breysse et al., J Neurosci, 23:8302-8309, 2003; and Wichmann and DeLong, Ann NY Acad Sci, 991:199-213, 2003). Overactivity or dysregulated activity of these nuclei contributes to the motor deficits of PD.
  • D 1 agonists enhance the release of GABA within the SNr, as measured by in vivo microdialysis (You et al., Neurosciece, 63:427-434, 1994; and Trevitt et al., Psychopharmacol, 159:229-237, 2002). Increased GABA would be expected to reduce SNr inhibition to the thalamus and increase motor activity.
  • Stimulation of D 2 receptors in the dopamine-denervated rat inhibits striatopallidal activity as a result of reducing glutamatergic activity from the cortex (Centonze et al., Clin Neurophysiol, 110:2006-2013, 1999).
  • a downstream consequence of this action is a reduction in glutamate release from the STN projections in the output nuclei, which can be measured by in vivo microdialysis in the SNr or EP (Biggs et al., Brain Res, 753:163-175, 1997; and Biggs and Starr, Neurosci Biobehav Rev, 21:497-504, 1997).
  • 6-OHDA 6-hydroxydopamine
  • 6-OHDA is used to selectively lesion dopamine neurons innervating the striatum on one side, thereby causing greater thalamic inhibition on the lesioned side
  • Agents that modify basal ganglia output manifest their effects as rotational behavior, the quantity and duration of which can be readily quantified. This model has also been used to assess the dyskinetic potential of drug treatments.
  • D 1 agonists and selective D 2 agonists have demonstrated efficacy in rodent and primate models of PD (Schwarting and Huston, supra 1996; and Rascol et al., Ann Neurol, 45:736-741, 1999).
  • L-DOPA and D 2 agonists have found widespread use in treating PD (Jenner, Curr Opin Neurol, 16(S1):S3-S7, 2003).
  • D 2 agonists have been utilized as monotherapy for mild or early stage PD and as adjuncts to L-DOPA in more advanced PD. If therapy is initiated with D 2 agonists alone, the incidence of dyskinesia is drastically reduced (Jenner, supra 2003).
  • D 2 agonists do not display the spectrum of efficacy seen with L-DOPA and as PD progresses L-DOPA therapy is frequently initiated (Jenner, supra 2003; and Lang and Lees, Mov Disord, 17(S4):S23-S37, 2002). Motor fluctuations and dyskinesias emerge as treatment with L-DOPA continues. It has been suggested that D 1 agonists may show a more favorable therapeutic profile than existing medications, but there has been little success with D 1 agonists for PD in the past (Emre et al., Mod Disord, 7:239-243, 1992; and Rascol et al., supra, 1999). There is a clear need to improve on drug therapy for PD.
  • the preferred dopamine receptor agonists for use with the present invention are partial agonists of the D1 dopamine receptor subtypes.
  • Particular examples of such compounds are benzazepine derivatives such as SKF38393, SKF83959, SKF81297, SKF77434, SKF75670, SKF82958 (Pettersson et al., J Med Chem, 33:2197-2204, 1990; and Neumeyer et al., Eur J Pharmacol, 474:137-140, 2003).
  • full agonists of the D1 receptor are used.
  • Examples of such compounds are tetrahydroisoquinoline derivatives such as dinapsoline (Ghosh et al., J Med Chem, 39:549-555, 1996), conformationally restricted phenanthridines such as dihydrexidine (Brewster et al., J Med Chem, 33:1756-1764, 1990), isochromans such as A68930, tetrahydroisoquinoline derivatives such as dinapsoline (Ghosh et al., J Med Chem, 39:549-555, 1996) and benzopyrans such as A-68930 and A-77636 (DeNinno et al., J Med Chem 33:2948-2950, 1990).
  • tetrahydroisoquinoline derivatives such as dinapsoline (Ghosh et al., J Med Chem, 39:549-555, 1996)
  • conformationally restricted phenanthridines such as dihydrexidine
  • full agonists of the D1 receptor are used at a significantly reduced dosage (e.g., at or below the threshold of clinical efficacy) as compared to the dosage used as monotherapy.
  • partial agonists of the D2 receptor subtype or full agonists of the D2 receptors are used (Jenner Curr Opin Neurol 16[Suppl 1]:S3-S7, 2003; Bonucelli Curr Opin Neurol 16[Suppl 1]:S13-S19, 2003), as described for D1 agonists.
  • the preferred serotonin (5-HT) receptor agonists for use with this invention are full agonists at the 5-HT1A receptor subtype, or partial agonists with moderate to high efficacy at the 5-HT1A receptor subtype.
  • Particular examples of such compounds include phenylpiperazines such as flesinoxan, eltoprazine, ipsapirone, gepirone, tandospirone, buspirone, eltoprazine, befiperide, BMY7378, flibanserin, and aripiprazole (See, e.g., van Steen et al., J Med Chem, 36:2751-2760, 1993; van Steen et al., J Med Chem, 37:2761-2773, 1994; van Steen et al., J Med Chem, 38:4303-4308, 1995; Kuipers et al., J Med Chem, 38:1942-1954, 1995; Kuipers et al., J Med Chem
  • Particular examples also include substituted chromane derivatives such as sarizotan, 8-OHDPAT, alnespirone, BAY X 3702 (De Vry et al., J Pharmacol Exp Ther, 284:1082-1094, 1998; and Hammarberg et al., J Med Chem, 43:2837-2850, 2000) and pyridinemethylamine derivatives (Vacher et al., J Med Chem, 42:1648-1660, 1999).
  • substituted chromane derivatives such as sarizotan, 8-OHDPAT, alnespirone, BAY X 3702 (De Vry et al., J Pharmacol Exp Ther, 284:1082-1094, 1998; and Hammarberg et al., J Med Chem, 43:2837-2850, 2000) and pyridinemethylamine derivatives (Vacher et al., J Med Chem, 42:1648-1660, 1999).
  • the preconditioning period refers to the length of time required for pretreatment with a 5-HT1A receptor agonist (in the presence or absence of a dopamine agonist) before an anti-Parkinsonian effect is observed in response to combined 5-HT1A agonist-dopamine agonist therapy. This period is hypothesized to reflect the time required for neural changes to occur in response to the repeated 5-HT1A agonist administrations. Nonetheless, an understanding of the mechanism is not necessary in order to make and use the present invention.
  • a simple assessment of levels of the 5-HT metabolite 5-hydroxyindoleacetic acid (5-HIAA) in blood taken at the onset of 5-HT1A agonist treatment and following repeated 5-HT1A agonist administration provides an indication as to whether the preconditioning period is adequate. For instance, a 20% or greater difference in 5-HIAA level between first and second sampling in which the 5-HIAA is lower at first sampling and the 5-HIAA level from the second sampling is anticipated to be in the range of 4.2-18.9 ng/ml (Yeung et al., J Pharm Sci, 85: 451-453, 1996) is contemplated to be indicative of sufficient preconditioning.
  • a comparison of the plasma levels of cortisol and change in body temperature in response to the first administration of drug versus responses following drug administration after several weeks of treatment is also contemplated to be useful for assessing whether the preconditioning period is sufficient for effecting an anti-Parkinsonian response to subsequent combined 5-HT1A agonist-dopamine agonist therapy.
  • an increase in plasma cortisol of 30% or greater and a reduction in body temperature of 0.2° C. or greater when assessed 90 minutes after drug intake is expected following initial 5-HT1A agonist treatment (Sargent et al., Psychopharmacol, 132: 296-302, 1997; and Yatham et al., J Affect Dis, 54:295-301, 1999). It is contemplated that a significant attenuation of these responses will be seen when assessed 90 minutes following drug intake in patients that have received a sufficient preconditioning period of treatment (Sargent et al., Psychopharmacol, 132: 296-302, 1997).
  • Dyskinesia scores were based on the following scale: 0—no evidence of abnormal activity; 1—mild, intermittent forelimb dyskinesia; 2—severe, more persistent forelimb dyskinesia associated with animal directing activity toward forelimb; 3-severe limb and axial dyskinesia associated with loss of balance and consistent “barrel-rolling” activity. The dyskinesia scores that were obtained were averaged to generate a dyskinesia index.
  • Rats that demonstrated contralateral turning indicative of a successful lesion received a daily subcutaneous injection of L-DOPA (10 mg/kg+6.25 mg/kg benserazide) or L-DOPA plus R(+)-8-OHDPAT (0.3 mg/kg). Ipsilateral turning as well as contralateral turning was recorded and quantified. As shown in FIG. 1 , L-DOPA elicited almost exclusively contralateral turning, which became progressively greater upon repeated daily dosing. This represents the “priming” phenomenon, which may reflect neurobiological changes associated with L-DOPA-induced dyskinesias as well as an anti-Parkinsonian response (Lane et al., Exp Neurol 197:284-290, 2006).
  • L-DOPA-treated animals Although not quantified in this experiment, the turning response of L-DOPA-treated animals was observed to become progressively more uncoordinated upon repeated L-DOPA administration, suggesting the emergence of dyskinetic activity. Despite this, L-DOPA-treated animals turned approximately 3-fold more on Day 15 than on Day 1. Animals treated with L-DOPA plus R(+)-8-OHDPAT displayed ipsilateral turning early in the observation period (first 30 min) and then exhibited contralateral turning. The ipsilateral turning could be elicited by R(+)-8-OHDPAT treatment alone. Even so, contralateral turning elicited by L-DOPA plus R(+)-8-OHDPAT on Day 15 was approximately 2-fold greater than the contralateral turning exhibited on Day 1.
  • a Serotonin Agonist Enhances the Anti-Parkinsonian Actions of Dopamine Agonists in a Rat Model of Parkinson's Disease
  • Such testing was carried out in a randomized fashion with different doses of dopamine agonist being tested every 4 th day (e.g. 3 days between dopamine agonist administration). Throughout this testing phase the control group continued to receive daily injection of vehicle and the 8-OHDPAT group continued to receive daily injection of 0.3 mg/kg R(+)-8-OHDPAT. On days when dopamine agonists were tested the vehicle or R(+)-8-OHDPAT was administered simultaneously with the dopamine agonist.
  • FIGS. 2 and 3 illustrate that administration of a 5-HT1A agonist enhances the anti-parkinsonian action of D1 and D2 agonists, as determined by quantitating contralateral turning in the hemi-lesioned 6-OHDA-treated rat.
  • animals were given the 5-HT1A agonist R(+)-8-OHDPAT (0.3 mg/kg) or vehicle (0.9% saline containing ascorbate) for 14 days.
  • Rats that received R(+)-8-OHDPAT for 14 days were then given R(+)-8-OHDPAT (0.3 mg/kg) plus a dopamine agonist on test day, and were observed for 3 hr for anti-parkinsonian effect (contralateral turning).
  • FIG. 2 depicts the data obtained when the D1 agonist SKF 38393 (0.1 mg/kg) was administered on test day. Animals that received R(+)-8-OHDPAT exhibited pronounced contralateral turning when SKF 38393 was administered. This turning behavior was significantly greater than what was observed for animals that received vehicle (p ⁇ 0.005). In fact, animals that received vehicle plus SKF 38393 displayed no contralateral turning.
  • FIG. 3 depicts the data obtained when the D2 agonist quinpirole (0.05 mg/kg) was administered on test day. Animals that received R(+)-8-OHDPAT exhibited significantly greater contralateral turning following quinpirole as compared to animals that received vehicle plus quinpirole (p ⁇ 0.001).
  • FIG. 10 depicts the rotational response on day 15 in rats given once daily administration of vehicle+SKF 38393 (0.1, 0.3 or 1.0 mg/kg). There is little net contralateral turning by day 15 of treatment. In contrast, animals receiving once daily R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 treatments displayed a significantly greater contralateral turning response that displayed a dose-dependency with respect to the dose of SKF administered.
  • FIG. 11 compares the rotational response between pre-screened (“primed”) and non-pre-screened (“unprimed”) rats after 15 days of once daily administration of R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 (0.3 mg/kg). There is no significant difference between the groups.
  • a Serotonin Agonist Reduces Dyskinetic Behavior Elicited by Moderate Doses of Dopamine Agonists
  • FIG. 5 illustrates that repeated R(+)-8-OHDPAT administration reduced dyskinetic behavior elicited by a moderately high dose of SKF 38393 (3.0 mg/kg).
  • FIG. 6 illustrates that repeated R(+)-8-OHDPAT administration reduced dyskinetic behavior elicited by quinpirole (0.05 mg/kg).
  • FIG. 7 illustrates that pretreatment of animals on test day with WAY 100635 (0.1 mg/kg) prior to receiving R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 (0.3 mg/kg) blocked the facilitating effects of R(+)-8-OHDPAT on the anti-parkinsonian action of SKF 38393.
  • ⁇ FosB is a truncated splice variant of FosB that represents a long-lived transcription factor.
  • ⁇ FosB has been shown to be upregulated following DA denervation in rodent and primate models of PD, as well as in PD patients (Cenci et al., Neuroscience, 94:515-527, 1999; Perez-Otano et al., Brain Res Mol Brain Res, 53:41-52 1998; and Tekumalla et al., Biol Psychiatry, 50:813-816, 2001).
  • FIG. 8 provides an image of a typical blot.
  • FIG. 9 illustrates that once daily administration of L-DOPA (25 mg/kg plus 6.25 mg/kg benserazide) or SKF 38393 (1 mg/kg) for 15 days elicits a significant increase in ⁇ FosB in the denervated striatum of unilateral 6-OHDA-lesioned rats (*, significantly greater than vehicle-treated; p ⁇ 0.05).
  • a Serotonin Agonist Enhances the Anti-Parkinsonian Actions of Dopamine Agonists in a Marmoset Model of Parkinson's Disease
  • This example describes testing a serotonin agonist with a dopamine agonist in a nonhuman primate model of Parkinson's disease.
  • adult common marmosets Callithrix jacchus ) received either 0.9% saline SC (1 ml/kg) daily for five consecutive days (na ⁇ ve controls) or MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride) SC (2 mg/kg) daily for five consecutive days.
  • MPTP administration induces a motor disorder characterized by akinesia, bradykinesia and rigidity, which is relatively stable after an initial recovery period. Abnormal posture, loss of vocalization, diminished blinking, incoordination and a tremor are also frequently observed.
  • Efficacy of administration of a serotonin type 1A receptor agonist and a dopamine receptor agonist in reducing at least one symptom of MPTP-induced motor disorder is measured by behavioral assessment as described (Pearce et al., Psychopharmacol, 142:51-60, 1999). Briefly, automated locomotor counts, behavioral observation and dyskinesia scoring are recorded. Automated locomotor counts are taken through the use of cages equipped with multiple infrared photocells. The number of light beam interruptions caused by movement is enumerated in 10-min intervals over a 2-12 h and recorded by computer. An initial period of 30-60 min is used for habituation to the cages prior to drug dosing and behavioral assessment.
  • the animals are also closely monitored by trained observers (e.g., a clinical neurologist with experience in movement disorders).
  • the degree of motor dysfunction is scored on a disability grading system; alertness (normal/0, abnormal/1); reaction to stimuli (normal/0, reduced/1, slow/2, absent/3); blinking (normal/0, abnormal/1); checking movements (present/0, reduced/1, absent/2); posture (normal/0, abnormal trunk/+1, tail/+1, limbs/+1, grossly abnormal/4); balance (normal/0, unstable/1, spontaneous falls/2); motility (normal/0, mild slowing/1, moderate bradykinesia/2, akinesia/3); vocalisation (normal/0, reduced/1, absent/2); tremor (absent/0, present/1) fur condition (normal/0, dirty/1). These values are summed to yield a disability score out of 20. Disability scores are generated from continuous observation at half hour intervals before and during peak drug effect.
  • Abnormal movements are described according to classically defined criteria: chorea (rapid random kicking limb movements); athetosis (sinuous writhing limb movements); dystonia (abnormal sustained posturing); and stereotypy(repetitive purposeless or semipurposive movement).
  • a Serotonin Agonist Enhances the Anti-Parkinsonian Actions of Dopamine Agonists in a Parkinson's Disease Patients
  • This example describes testing a serotonin agonist with a dopamine agonist in a human Parkinson's disease patients. Briefly, all patients admitted to this study are diagnosed with idiopathic Parkinson's disease (See, e.g., Gibb and Lees, J Neurol Neurosurg Psychiatry, 51:745-752, 1988).
  • Efficacy of administration of a serotonin type 1A receptor agonist and a dopamine receptor agonist in reducing at least one symptom of Parkinson's disease is measured using the United Parkinson's Disease Rating Scale (Fahn et al. (eds.), Recent Developments in Parkinson's Disease, vol. 2, New York, N.Y.: Macmillan Publishing Co. Inc., pp. 153-163 and 293-304, 1987), whereas dyskinesias are assessed using an art recognized severity scale (Marconi et al., Mov Disord, 9:2-12, 1994). Briefly, the dyskinesia severity scale rates abnormal movements from 0 (none) to 4 (severe with markedly impaired function) in six different parts of the body (face, neck and trunk, and four limbs).

Abstract

The present invention relates to the co-administration of two neurotransmitter agonists to patients with motor disorders, for the purpose of symptom reduction. In particular the present invention provides methods and compositions for alleviation of akinesia, rigidity and/or tremor associated with Parkinson's disease.

Description

  • This application claims benefit of U.S. Provisional Application No. 60/777,939, filed on Feb. 28, 2006, herein incorporated by reference in its entirety.
  • This invention was made in part through funds from the Department of Veterans Affairs. As such, the United States government has certain rights in the invention.
  • FIELD OF THE INVENTION
  • The present invention relates to the co-administration of two neurotransmitter agonists to patients with motor disorders, for the purpose of symptom reduction. In particular the present invention provides methods and compositions for alleviation of akinesia, rigidity and/or tremor associated with Parkinson's disease.
  • BACKGROUND OF THE INVENTION
  • Parkinson's disease is a chronic, progressive, hypokinetic disorder characterized by impaired voluntary movement (See, Dale and Federman (eds.), WebMD Scientific American Medicine, NY: WebMD Corporation, Chapter 11, Section 15, pp. 1-21, 2001; Lang and Lozano, N Engl J Med, 339:1044, 1998; and Lang and Lozano, N Engl J Med, 339:1130, 1998). Parkinson's disease occurs as a result of the death of dopamine-producing neurons in the substantia nigra of the midbrain. Dopamine is a neurotransmitter, or chemical messenger, that transports signals to the parts of the brain that control movement initiation and coordination. The loss of dopamine in the brain is associated with multiple primary symptoms including: tremor of the hands, arms, legs, jaw, and face; rigidity or stiffness of the limbs and trunk; bradykinesia or slowness of movement; and postural instability or impaired balance and coordination.
  • Parkinson's disease afflicts more than one million persons in the United States alone (Lang and Lozano, supra, 1998), with approximately 50,000 new cases diagnosed each year. It is generally a disease of late middle age, with typical onset occurring at about age 60. About five percent of patients, however, have early-onset disease and are younger than 40 when symptoms begin.
  • Most current treatment strategies for Parkinson's disease (PD) focus on symptom control through one or more of medication, surgery, and physical therapy. The dopamine precursor, levodopa (L-DOPA) is still considered to be the gold standard in terms of treatment for PD (Schapira and Olanow, JAMA, 291:358-364, 2004). Unfortunately, L-DOPA can cause debilitating side effects (LeWitt and Nyholm Neurology, 62:S9-S16, 2004), including severe nausea, vomiting, and psychosis. Moreover, with prolonged use, patients frequently experience other side effects such as dyskinesias (spontaneous, involuntary movements) and “on-off” periods when the medication will suddenly start or stop working.
  • Surgical treatments are considered for patients with advanced disease or who have not responded adequately to medications. Accepted surgical treatments involve either the creation of small, precise lesions or the implantation of stimulating electrodes in specific brain regions that appear to be overactive in Parkinson's disease. More recently, implantation of fetal dopaminergic tissue has been utilized as a means of restoring dopamine levels in the brains of patients with Parkinson's disease (See, e.g., Freed et al., Arch Neurol, 47:505-512, 1990; and Lindvall et al., Science, 247:574-577, 1990). These types of methods, however, are highly invasive.
  • Thus, what is needed in the art are noninvasive treatment strategies for effectively controlling symptoms of Parkinson's disease and other movement disorders. In addition, it would be desirable to be in possession of therapy regimens that maximize the efficacy of existing medicines.
  • SUMMARY OF THE INVENTION
  • The present invention relates to the co-administration of two neurotransmitter agonists to patients with motor disorders, for the purpose of symptom reduction. In particular the present invention provides methods and compositions for alleviation of akinesia, rigidity and/or tremor associated with Parkinson's disease, as well as for alleviation of side effects associated with treatment associated with Parkinson's disease.
  • The present invention provides methods of treating a motor disorder, comprising: providing: i) a serotonin type 1A (5-HT1A) receptor agonist, ii) a dopamine receptor agonist, and iii) a subject diagnosed as having a motor disorder; and b) administering the serotonin type 1A receptor (5-HT1A) agonist to the subject during a pre-conditioning period; and c) co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with a low dose of the dopamine receptor agonist to the subject, under conditions suitable for reducing at least one symptom of the motor disorder, wherein the low dose of the dopamine receptor agonist is below that required for reducing at least one symptom of the motor disorder when administered in the absence of the serotonin type 1A (5-HT1A) receptor agonist. In some embodiments, the motor disorder is Parkinson's disease. In other embodiments, the motor disorder is parkinsonism arising from viral infection or drug use. In some preferred embodiments, the dopamine receptor agonist is not a dopamine precursor. In some embodiments, the serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of arylpiperazines, azapirones, aminotetralins, and aminochromanes. In other embodiments, the serotonin type 1A (5-HT1A) receptor agonist is 8-hydroxy-2-(di-n-propylamino)tetralin (8OH-DPAT). In some preferred embodiments, 8OH-DPAT is administered via a transdermal patch. In some embodiments, the serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of tandospirone, ipsapirone, gepirone, flibanserin, flesinoxan, sarizotan, repinotan and PRX-00023. In some preferred embodiments, the dopamine receptor agonist is a D1 type dopamine receptor agonist. In a subset of these embodiments, the D1 type dopamine receptor agonist is selected from the group consisting of SKF38393, SKF83959, SKF81297, SKF77434, SKF75670, SKF82958, and derivatives thereof. In further embodiments, the D1 type dopamine receptor agonist is selected from the group consisting of dihydrexidine, dinapsoline, A-77636, ABT-431, CY208-243, and A-68930. In other embodiments, the dopamine receptor agonist is a D2 type dopamine receptor agonist. In some of these embodiments, the D2 type dopamine receptor agonist is quinpirole. In other embodiments, the D2 type dopamine receptor agonist is selected from the group consisting of quinelorane, terguride, pergolide, talipexole, pramipexole, sumanirole, ropinirole, roxindole, bromocriptine, and cabergoline. In still further embodiments, the dopamine agonist is a dopamine precursor. In a subset of these embodiments, the dopamine precursor is levodopa (L-DOPA), which is administered with one or both of a L-aromatic amino acid decarboxylase inhibitor (e.g., carbidopa, benserazide, difluromethyldopa, a-methyldopa, etc.) and a catechol-O-methyltransferase (COMT) inhibitor (e.g., tolcapone and entacapone). In still further embodiments one or both of the serotonin type 1A (5-HT1A) receptor agonist and the dopamine receptor agonist is a D1 type dopamine receptor agonist. In a subset of these embodiments, the serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of sunipetron and aripiprazole. Also provided are methods further comprising administering a low dose of the dopamine receptor agonist to the subject during the pre-conditioning period, wherein the low dose is of the dopamine receptor agonist is below that required for reducing at least one symptom of the motor disorder when administered in the absence of the serotonin type 1A (5-HT1A) receptor agonist.
  • In some preferred embodiments, the present invention provides methods of treating Parkinson's disease, comprising: providing: i) a serotonin type 1A (5-HT1A) receptor agonist, ii) a dopamine receptor agonist, and iii) a subject diagnosed as having Parkinson's disease; and b) administering the serotonin type 1A receptor (5-HT1A) agonist to the subject during a pre-conditioning period; and c) co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with a low dose of the dopamine receptor agonist to the subject, under conditions suitable for reducing at least one symptom of Parkinson's disease, wherein the low dose of the dopamine receptor agonist is below that required for reducing at least one symptom of Parkinson's disease when administered in the absence of the serotonin type 1A (5-HT1A) receptor agonist. In some particularly preferred embodiments, the at least one symptom comprises one or more of bradykinesia, tremor, and muscle rigidity. Also provided are embodiments in which the co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with the dopamine receptor agonist to the subject is under conditions further suitable for reducing at least one side effect of the dopamine receptor agonist. In some preferred embodiments, the at least one side effect of the dopamine receptor agonist comprises dyskinesia (e.g., one or more of chewing, gnawing, twisting, tongue or mouth movements, head bobbing, and movements of the feet, hands, or shoulder). In other embodiments, the at least one side effect of the dopamine receptor agonist comprises a psychiatric disturbance (e.g., one or more of memory loss, anxiety, nervousness, agitation, restlessness, confusion, inability to sleep, nightmares, daytime tiredness, mental depression and euphoria). In some preferred embodiments, the dopamine receptor agonist is not a dopamine precursor.
  • Moreover, the present invention provides methods for treating a subject, comprising: providing; i) a serotonin type 1A (5-HT1A) receptor agonist, ii) a dopamine receptor agonist, iii) a subject receiving the dopamine receptor agonist for treatment of Parkinson's disease, wherein the subject is suffering from dopamine receptor agonist-induced dyskinesia; co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with the dopamine receptor agonist to the subject during a preconditioning period under conditions suitable for reducing the dopamine receptor agonist-induced dyskinesia; co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with the dopamine receptor agonist to the subject during a dosage optimization period, wherein dosage of the dopamine receptor agonist is gradually reduced until at least one symptom of the Parkinson's disease is re-emerges; and co-administering the serotonin type 1A receptor (5-HT1A) agonist in combination with the dopamine receptor agonist to the subject under conditions suitable for reducing the at least one symptom of the Parkinson's disease, while reducing the dopamine receptor agonist-induced dyskinesia, wherein dosage of the dopamine receptor agonist is below that of step b and above that of step c.
  • In addition, the present invention provides kits for treating Parkinson's disease comprising a serotonin type 1A (5-HT1A) receptor agonist, a dopamine receptor agonist, and instructions for administering the serotonin type 1A (5-HT1A) receptor agonist and the dopamine receptor agonist to a subject diagnosed as having Parkinson's disease, wherein the dopamine receptor agonist is administered at a dose below that required for reducing at least one symptom of the Parkinson's disease when administered in the absence of the serotonin type 1A (5-HT1A) receptor agonist. In some embodiments, the kit comprises a package (e.g., blister pack). In some preferred embodiments, the dopamine receptor agonist is not a dopamine precursor.
  • Also provided are compositions for treating Parkinson's disease comprising: a therapeutic dose of a serotonin type 1A (5-HT1A) receptor agonist and a subtherapeutic dose of a dopamine receptor agonist. In some preferred embodiments, the dopamine receptor agonist is not a dopamine precursor. In some particularly preferred embodiments, the compositions further comprise a pharmaceutically acceptable excipient.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts the effects of repeated daily dosing with L-DOPA (10 mg/kg+6.25 mg/kg benserazide) or L-DOPA plus R(+)-8-OHDPAT (0.3 mg/kg). Ipsilateral turning as well as contralateral turning was recorded and quantified.
  • FIG. 2 demonstrates that a 5-HT1A agonist enhances the anti-parkinsonian action of a D1 type dopamine agonist as demonstrated by contralateral turning in the unilateral 6-OHDA-lesioned rat. The 5-HT1A agonist R(+)-8-OHDPAT (0.3 mg/kg) or vehicle (0.9% saline containing ascorbate) was administered for 14 days. On Day 15, rats were given either vehicle or R(+)-8-OHDPAT (0.3 mg/kg), plus a low dose D1 agonist SKF 38393 (0.1 mg/kg) and observed for 3 hr.
  • FIG. 3 demonstrates that a 5-HT1A agonist enhances the anti-parkinsonian action of a D2 type dopamine agonist as demonstrated by contralateral turning in the unilateral 6-OHDA-lesioned rat. The 5-HT1A agonist R(+)-8-OHDPAT (0.3 mg/kg) or vehicle (0.9% saline containing ascorbate) was administered for 14 days. On Day 15, rats were given either vehicle or R(+)-8-OHDPAT (0.3 mg/kg), plus a low dose D2 agonist quinpirole (0.05 mg/kg) and observed for 3 hr.
  • FIG. 4 illustrates that both repeated and concomitant administration of a serotonin agonist was required to enhance that anti-parkinsonian actions of a dopamine agonist.
  • FIG. 5 demonstrates that repeated administration of R(+)-8-OHDPAT reduced dyskinetic behavior elicited by a moderately high dose of the D1 agonist SKF 38393 (3.0 mg/kg).
  • FIG. 6 demonstrates that repeated administration of R(+)-8-OHDPAT reduced dyskinetic behavior elicited by a moderately high dose of the D2 agonist quinpirole (0.05 mg/kg).
  • FIG. 7 illustrates that pre-treatment with a selective 5-HT1A antagonist (WAY 100635) blocked the facilitating effects of a serotonin agonist on the anti-parkinsonian action of a dopamine agonist.
  • FIG. 8 is an image of an immunoblot of striatal extracts probed with an anti-Fos antibody.
  • FIG. 9 illustrates that once daily administration of L-DOPA (25 mg/kg plus 6.25 mg/kg benserazide) or SKF 38393 (1 mg/kg) for 15 days elicits a significant increase in ΔFosB in the denervated striatum of unilateral 6-OHDA-lesioned rats (*, significantly greater than vehicle-treated; p<0.05). The concomitant administration of the 5-HT1A agonist R(+)-8-OHDPAT (0.3 mg/kg) with SKF 38393 prevents the D1 agonist-induced rise in ΔFosB (#, significantly less than VEH+SKF; p<0.05 and not significantly different from vehicle-treated).
  • FIG. 10 depicts the rotational response in rats given once daily administration of vehicle+SKF 38393 (0.1, 0.3 or 1.0 mg/kg) for 15 days. Animals receiving once daily R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 displayed a significantly greater contralateral turning response that displayed a dose-dependency with respect to the dose of SKF administered.
  • FIG. 11 compares the rotational response between pre-screened (“primed”) and non-pre-screened (“unprimed”) rats after 15 days of once daily administration of R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 (0.3 mg/kg). There is no significant difference between the groups.
  • DEFINITIONS
  • To facilitate an understanding of the present invention, a number of terms and phrases are defined below:
  • The terms “Parkinson's disease,” “Parkinson's” and “PD” refer to a neurological syndrome characterized by a dopamine deficiency, resulting from degenerative, vascular, or inflammatory changes in the basal ganglia of the substantia nigra. This term also refers to a syndrome which resembles Parkinson's disease, but which may or may not be caused by Parkinson's disease, such as Parkinsonian-like side effects caused by certain antipsychotic drugs. Parkinson's disease is also referred to as paralysis agitans and shaking palsy.
  • As used herein, the term “early stage of Parkinson's disease” refers broadly to the first stages in Parkinson's disease, wherein a person suffering from the disease exhibits mild symptoms that are not disabling, such as an episodic tremor of a single limb (e.g., the hand), and which affect only one side of the body.
  • In contrast, the term “advanced stage of Parkinson's disease” refers broadly to a more progressive stage in Parkinson's disease, wherein a person suffering from the disease exhibits symptoms which are typically severe and which may lead to some disability (e.g., tremors encompassing both sides of the body, balance problems, etc.). Symptoms associated with advanced stage Parkinson's disease may vary significantly in individuals, and may take many years to manifest after the initial appearance of the disease.
  • The terms “subject suffering from a Parkinson's disease” and “subject suffering from parkinsonism” as used herein, refer to both humans and animals displaying one or more symptom of Parkinson's disease. The primary symptoms of Parkinson's disease include bradykinesia, tremor, and muscle rigidity. The term “animals” refers to all non-human animals. Such non-human animals include, but are not limited to, vertebrates such as rodents, non-human primates, ovines, bovines, lagomorphs, porcines, caprines, equines, canines, felines, aves, etc.
  • The terms “dopamine,” “DA” and “4-(2-aminoethyl)benzene-1,2-diol,” refer to a catecholamine neurotransmitter and hormone, formed first by the hydroxylation of the amino acid tyrosine to dihydroxyphenylalanine (dopa), and then by decarboxylation of dihydroxyphenylalanine (dopa) to dopamine. Dopamine is a precursor of adrenaline (epinephrine) and noradrenaline (norepinephrine).
  • The terms “serotonin,” “5-hydroxytryptamine” and “5-HT” refers to a phenolic amine neurotransmitter produced from tryptophan by hydroxylation and decarboxylation in serotonergic neurons of the central nervous system and enterochromaffin cells of the gastrointestinal tract. Serotonin is a precursor of melatonin.
  • As used herein, the term “symptoms” refers to the outward manifestations of a disease. For instance, patients with Parkinson's disease exhibit symptoms such as bradykinesia, tremor, and muscle rigidity. Additional symptoms but are not limited to atypical gait, postural instability, and loss of balance.
  • The term “alleviating” refers to the act of providing relief from some painful state. As used herein, the term “alleviating” refers to the lessoning of symptoms of Parkinson's disease or to the lessoning of side effects associated with long term L-dopa administration. In some embodiments, the term “alleviating” comprises curing Parkinson's disease. In other embodiments, the term “alleviating” comprises decreasing the severity of the symptoms of Parkinson's disease as measured by any one of several accepted diagnostic measures including but not limited to UPDRS, Schwab and England Scale, and CAPIT.
  • The terms “Unified Parkinson's Disease Rating Scale” and “UPDRS” refer to a standardized tool used to measure Parkinson's Disease severity, as described by Fahn et al., in Recent Developments in Parkinson's Disease, Fahn et al. (eds.) Plurham Park, N.J.: Macmillian Healthcare Information, 2:153-163, 1987.
  • “The term “Schwab and England Scale” refers to a standardized tool used to measure the efficacy of surgical treatment for Parkinson's Disease, as described by Schwab and England, in Third Symposium on Parkinson's Disease, Gillingham and Donaldon (eds.) Edinburgh, Scotland: Livingstone, pp. 152-157, 1969.
  • The terms “Core Assessment Program for Intracerebral Transplantation” and “CAPIT” refer to a standardized tool used to measure the efficacy of cerebral surgery, as described by Langston et al., Mov Disord, 7:2-13, 1992.
  • The terms “agonist” and “stimulator” refers to molecules or compounds that mimic the action of a “native” or “natural” compound. Agonists may be homologous to these natural compounds in respect to conformation, charge or other characteristics. Thus, agonists may be recognized by receptors expressed on cell surfaces. This recognition may result in physiologic and/or biochemical changes within the cell, such that the cell reacts to the presence of the agonist in the same manner as if the natural compound was present. The term “agonist” also encompasses the class of agents known as partial agonists, in which the maximal physiologic and/or biochemical changes that occur upon partial agonist administration may be less than the maximal effect exhibited by full agonists or the natural compound. In preferred embodiments, the term agonist refers to molecules that bind to and activate one or both of serotonin type 1A and dopamine type receptors.
  • As used herein, the terms “antagonist” and “inhibitor” refer to molecules or compounds that inhibit the action of a “native” or “natural” compound. Antagonists may or may not be homologous to these natural compounds in respect to conformation, charge or other characteristics. Thus, antagonists may be recognized by the same or different receptors than are recognized by an agonist. Antagonists may have allosteric effects, which prevent the action of an agonist. In contrast to the agonists, antagonistic compounds do not result in physiologic and/or biochemical changes within the cell, such that the cell reacts to the presence of the antagonist in the same manner as if the natural compound was absent.
  • As used herein the term “subtherapeutic dose” refers to a quantity of a neurotransmitter agonist (e.g., D1 agonist) below that used to treat disease when used as a monotherapy. In an exemplary embodiment, a subtherapeutic dose of a D1 agonist is equivalent to 5 mg of SKF38393 administered to an adult human subject four times a day.
  • As used herein, the term “therapeutic dose” refers to a quantity of a neurotransmitter agonist (e.g., 5-HT1A agonist) used to treat disease (e.g., effect the cure of or that will correct the manifestations of a disease). In an exemplary embodiment, a therapeutic dose of a 5-HT1A agonist is equivalent to 5 mg of sarizotan administered to an adult human subject two or three times per day. In another exemplary embodiment, a therapeutic dose of a 5-HT1A agonist is equivalent to 5 mg of ipsapirone administered to an adult human subject four times a day.
  • DESCRIPTION OF THE INVENTION
  • The present invention relates to the co-administration of two neurotransmitter agonists to patients with motor disorders, for the purpose of symptom reduction. In particular the present invention provides methods and compositions for alleviation of akinesia, rigidity and/or tremor associated with Parkinson's disease.
  • Neurotransmitters
  • Parkinson's disease (PD) and related neurological disorders (e.g. postencephalitis parkinsonism, Hallervorden-Soatz disease) involve the progressive degeneration of dopamine neurons innervating the striatum. The loss of dopamine leads to excessive inhibition of thalamocortical motor activity, for which the primary manifestations are rigidity, bradykinesia, tremor and postural instability (Fahn, Ann NY Acad Sci, 991:1-14, 2003; and Hamani and Lozano, Ann NY Acad Sci, 991:15-21, 2003). The striatum is a major input structure that receives dense dopaminergic projections from the substantia nigra pars compacta and glutamatergic projections from the cortex and thalamus. Two efferent pathways originate in the striatum and utilize the inhibitory transmitter gamma-aminobutyric acid (GABA). The direct pathway is comprised of medium spiny striatonigral neurons that project directly to the output nuclei of the basal ganglia, which are the internal globus pallidus (rat homologue is the entopeduncular nucleus or EP) and the substantia nigra pars reticulata (SNr). These striatonigral neurons predominantly express dopamine D1 receptors and co-express dynorphin and substance P (Gerfen, Clin Neuropharmacol, 18:S162-S177, 1995). The indirect pathway is comprised of medium spiny striatopallidal neurons that project to the external globus pallidus (GP). These striatopallidal neurons predominantly express dopamine D2 receptors and co-express enkephalin (Gerfen, supra, 1995). The GP sends GABA projections to the subthalamic nucleus (STN), which in turn, sends glutamate projections to the EP and SNr. The EP and SNr serve as the final output structures of the basal ganglia and innervate the motor thalamus with inhibitory GABA projections. In PD and pre-clinical models of PD there is evidence of de-synchronization and hyperactivity in the STN, EP and SNr (Breysse et al., J Neurosci, 23:8302-8309, 2003; and Wichmann and DeLong, Ann NY Acad Sci, 991:199-213, 2003). Overactivity or dysregulated activity of these nuclei contributes to the motor deficits of PD.
  • Dopamine Replacement Therapy
  • The effects of D1 agonists on striatonigral activity are somewhat controversial (Calabresi et al., Neurosci Biobehav Rev, 21:519-523, 1997). However, in the dopamine-denervated rat, D1 agonists enhance the release of GABA within the SNr, as measured by in vivo microdialysis (You et al., Neurosciece, 63:427-434, 1994; and Trevitt et al., Psychopharmacol, 159:229-237, 2002). Increased GABA would be expected to reduce SNr inhibition to the thalamus and increase motor activity. Stimulation of D2 receptors in the dopamine-denervated rat inhibits striatopallidal activity as a result of reducing glutamatergic activity from the cortex (Centonze et al., Clin Neurophysiol, 110:2006-2013, 1999). A downstream consequence of this action is a reduction in glutamate release from the STN projections in the output nuclei, which can be measured by in vivo microdialysis in the SNr or EP (Biggs et al., Brain Res, 753:163-175, 1997; and Biggs and Starr, Neurosci Biobehav Rev, 21:497-504, 1997). These techniques are contemplated to be useful for monitoring the effects on excitatory and inhibitory transmissions in the output nuclei of the anti-parkinsonian therapies of the present invention.
  • In the unilateral 6-hydroxydopamine (6-OHDA)-lesioned rat model of PD, 6-OHDA is used to selectively lesion dopamine neurons innervating the striatum on one side, thereby causing greater thalamic inhibition on the lesioned side (Schwarting and Huston, Prog Neurobiol, 50:275-331, 1996; and Deumens et al., Exp Neurol, 175:303-317, 2002). Agents that modify basal ganglia output manifest their effects as rotational behavior, the quantity and duration of which can be readily quantified. This model has also been used to assess the dyskinetic potential of drug treatments. Along these lines, a progressive increase in rotational response is observed upon repeated daily treatment with DA agonists or L-DOPA. This augmentation of behavior is often referred to as “priming” and has been taken as an index of dyskinetic potential, although there is currently considerable debate as to whether “priming” is exclusively indicative of motor complications (Di Chiara et al., Dev Pharmacol Ther, 18:223-227, 1992; and Henry et al., Exp Neurol, 151:334-342. 1998; Lane et al., Exp Neurol, 197:284-290, 2006; Marin et al., Exp Neurol, 197:269-274, 2006). Numerous studies have used this phenomenon to establish associations between the priming effect and neurobiological changes.
  • Selective D1 agonists and selective D2 agonists have demonstrated efficacy in rodent and primate models of PD (Schwarting and Huston, supra 1996; and Rascol et al., Ann Neurol, 45:736-741, 1999). However, only L-DOPA and D2 agonists have found widespread use in treating PD (Jenner, Curr Opin Neurol, 16(S1):S3-S7, 2003). D2 agonists have been utilized as monotherapy for mild or early stage PD and as adjuncts to L-DOPA in more advanced PD. If therapy is initiated with D2 agonists alone, the incidence of dyskinesia is drastically reduced (Jenner, supra 2003). Unfortunately, D2 agonists do not display the spectrum of efficacy seen with L-DOPA and as PD progresses L-DOPA therapy is frequently initiated (Jenner, supra 2003; and Lang and Lees, Mov Disord, 17(S4):S23-S37, 2002). Motor fluctuations and dyskinesias emerge as treatment with L-DOPA continues. It has been suggested that D1 agonists may show a more favorable therapeutic profile than existing medications, but there has been little success with D1 agonists for PD in the past (Emre et al., Mod Disord, 7:239-243, 1992; and Rascol et al., supra, 1999). There is a clear need to improve on drug therapy for PD.
  • The preferred dopamine receptor agonists for use with the present invention are partial agonists of the D1 dopamine receptor subtypes. Particular examples of such compounds are benzazepine derivatives such as SKF38393, SKF83959, SKF81297, SKF77434, SKF75670, SKF82958 (Pettersson et al., J Med Chem, 33:2197-2204, 1990; and Neumeyer et al., Eur J Pharmacol, 474:137-140, 2003). In some particularly preferred embodiments when given alone their anti-Parkinsonian action would be minimal or not evident. In further embodiments, full agonists of the D1 receptor are used. Examples of such compounds are tetrahydroisoquinoline derivatives such as dinapsoline (Ghosh et al., J Med Chem, 39:549-555, 1996), conformationally restricted phenanthridines such as dihydrexidine (Brewster et al., J Med Chem, 33:1756-1764, 1990), isochromans such as A68930, tetrahydroisoquinoline derivatives such as dinapsoline (Ghosh et al., J Med Chem, 39:549-555, 1996) and benzopyrans such as A-68930 and A-77636 (DeNinno et al., J Med Chem 33:2948-2950, 1990). However, in preferred embodiments full agonists of the D1 receptor are used at a significantly reduced dosage (e.g., at or below the threshold of clinical efficacy) as compared to the dosage used as monotherapy. In further embodiments, partial agonists of the D2 receptor subtype or full agonists of the D2 receptors are used (Jenner Curr Opin Neurol 16[Suppl 1]:S3-S7, 2003; Bonucelli Curr Opin Neurol 16[Suppl 1]:S13-S19, 2003), as described for D1 agonists.
  • Serotonin Adjuvant Therapy
  • The preferred serotonin (5-HT) receptor agonists for use with this invention are full agonists at the 5-HT1A receptor subtype, or partial agonists with moderate to high efficacy at the 5-HT1A receptor subtype. Particular examples of such compounds include phenylpiperazines such as flesinoxan, eltoprazine, ipsapirone, gepirone, tandospirone, buspirone, eltoprazine, befiperide, BMY7378, flibanserin, and aripiprazole (See, e.g., van Steen et al., J Med Chem, 36:2751-2760, 1993; van Steen et al., J Med Chem, 37:2761-2773, 1994; van Steen et al., J Med Chem, 38:4303-4308, 1995; Kuipers et al., J Med Chem, 38:1942-1954, 1995; Kuipers et al., J Med Chem, 40:300-312, 1997; and Heinrich et al., J Med Chem, 47:4684-4692, 2004). Particular examples also include substituted chromane derivatives such as sarizotan, 8-OHDPAT, alnespirone, BAY X 3702 (De Vry et al., J Pharmacol Exp Ther, 284:1082-1094, 1998; and Hammarberg et al., J Med Chem, 43:2837-2850, 2000) and pyridinemethylamine derivatives (Vacher et al., J Med Chem, 42:1648-1660, 1999).
  • Serotonin Preconditioning Period
  • The preconditioning period refers to the length of time required for pretreatment with a 5-HT1A receptor agonist (in the presence or absence of a dopamine agonist) before an anti-Parkinsonian effect is observed in response to combined 5-HT1A agonist-dopamine agonist therapy. This period is hypothesized to reflect the time required for neural changes to occur in response to the repeated 5-HT1A agonist administrations. Nonetheless, an understanding of the mechanism is not necessary in order to make and use the present invention. It is contemplated that a simple assessment of levels of the 5-HT metabolite 5-hydroxyindoleacetic acid (5-HIAA) in blood taken at the onset of 5-HT1A agonist treatment and following repeated 5-HT1A agonist administration (e.g., days to weeks) provides an indication as to whether the preconditioning period is adequate. For instance, a 20% or greater difference in 5-HIAA level between first and second sampling in which the 5-HIAA is lower at first sampling and the 5-HIAA level from the second sampling is anticipated to be in the range of 4.2-18.9 ng/ml (Yeung et al., J Pharm Sci, 85: 451-453, 1996) is contemplated to be indicative of sufficient preconditioning. Alternatively, a comparison of the plasma levels of cortisol and change in body temperature in response to the first administration of drug versus responses following drug administration after several weeks of treatment is also contemplated to be useful for assessing whether the preconditioning period is sufficient for effecting an anti-Parkinsonian response to subsequent combined 5-HT1A agonist-dopamine agonist therapy. For instance, an increase in plasma cortisol of 30% or greater and a reduction in body temperature of 0.2° C. or greater when assessed 90 minutes after drug intake is expected following initial 5-HT1A agonist treatment (Sargent et al., Psychopharmacol, 132: 296-302, 1997; and Yatham et al., J Affect Dis, 54:295-301, 1999). It is contemplated that a significant attenuation of these responses will be seen when assessed 90 minutes following drug intake in patients that have received a sufficient preconditioning period of treatment (Sargent et al., Psychopharmacol, 132: 296-302, 1997).
  • Experimental
  • The following examples are provided in order to demonstrate and further illustrate certain preferred embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof.
  • In the experimental disclosure which follows, the following abbreviations apply: U (units); N (normal); M (molar); mM (millimolar); μM (micromolar); mol (moles); mmol (millimoles); μmol (micromoles); nmol (nanomoles); pmol (picomoles); g (grams); mg (milligrams); μg (micrograms); ng (nanograms); l or L (liters); ml (milliliters); μl (microliters); cm (centimeters); mm (millimeters); μm (micrometers); nm (nanometers); ° C. (degrees Centigrade); OD (optical density); bp (base pairs); PCR (polymerase chain reaction); RT (reverse transcriptase); DMEM (Dulbecco's modified Eagle medium); EDTA (ethylenediaminetetraacetic acid); FBS (fetal bovine serum); L-DOPA (levodopa); 6-OHDA (6-hydroxydopamine); and PBS (phosphate buffered saline).
  • EXAMPLE 1 Motor Effects of Co-Administration of a Serotonin Agonist and L-DOPA in a Rat Model of Parkinson's Disease
  • Preparation of hemi-lesioned 6-hydroxydopamine (6-OHDA)-treated rat model of Parkinson 's disease. Rats were rendered parkinsonian on one side of the brain by unilateral destruction of nigrostriatal dopamine neurons using the selective neurotoxin 6-OHDA and stereotaxic procedures. Animals were allowed to recover for at least 2 weeks following this procedure. Animals were then screened for efficacy of the lesion by subcutaneous injection of L-DOPA (25 mg/kg) plus the decarboxylase inhibitor benserazide (6.25 mg/kg) followed by assessment of motor activity.
  • Assessment of motor activity. Anti-parkinsonian action in the hemi-lesioned 6-OHDA-treated rat model was assessed by the elicitation of rotational activity to the side opposite the lesioned hemisphere (contralateral turns) following drug treatment. The number of contralateral turns was assessed by videotape recording of animals and counting the number of turns.
  • Assessment of dyskinetic activity. Observer assessment of motor dyskinesias was carried out be viewing the videotaped recording of rats during testing for rotational activity. A dyskinesia score was generated at 10 minute intervals throughout the course of the rotational test (generally 3 hr). Dyskinesia scores were based on the following scale: 0—no evidence of abnormal activity; 1—mild, intermittent forelimb dyskinesia; 2—severe, more persistent forelimb dyskinesia associated with animal directing activity toward forelimb; 3-severe limb and axial dyskinesia associated with loss of balance and consistent “barrel-rolling” activity. The dyskinesia scores that were obtained were averaged to generate a dyskinesia index.
  • Treatment and Results. Rats that demonstrated contralateral turning indicative of a successful lesion received a daily subcutaneous injection of L-DOPA (10 mg/kg+6.25 mg/kg benserazide) or L-DOPA plus R(+)-8-OHDPAT (0.3 mg/kg). Ipsilateral turning as well as contralateral turning was recorded and quantified. As shown in FIG. 1, L-DOPA elicited almost exclusively contralateral turning, which became progressively greater upon repeated daily dosing. This represents the “priming” phenomenon, which may reflect neurobiological changes associated with L-DOPA-induced dyskinesias as well as an anti-Parkinsonian response (Lane et al., Exp Neurol 197:284-290, 2006). Although not quantified in this experiment, the turning response of L-DOPA-treated animals was observed to become progressively more uncoordinated upon repeated L-DOPA administration, suggesting the emergence of dyskinetic activity. Despite this, L-DOPA-treated animals turned approximately 3-fold more on Day 15 than on Day 1. Animals treated with L-DOPA plus R(+)-8-OHDPAT displayed ipsilateral turning early in the observation period (first 30 min) and then exhibited contralateral turning. The ipsilateral turning could be elicited by R(+)-8-OHDPAT treatment alone. Even so, contralateral turning elicited by L-DOPA plus R(+)-8-OHDPAT on Day 15 was approximately 2-fold greater than the contralateral turning exhibited on Day 1. Given that augmented turning can be used as an index of dyskinesia, the inventors cannot discount the possibility that rats receiving the combined treatment had displayed evidence of dyskinetic potential. However the inventors also noted that rats receiving the combined treatment turned in a much more natural, coordinated fashion. These observations are in line with a published report of reduced motor complications of levodopa treatment upon coadministration of the 5-HT1A agonist sarizotan (Bibbiani et al., Neurology, 57:1829-1834, 2001).
  • EXAMPLE 2 A Serotonin Agonist Enhances the Anti-Parkinsonian Actions of Dopamine Agonists in a Rat Model of Parkinson's Disease
  • Treatment. Hemi-lesioned 6-OHDA-treated rat that demonstrated contralateral turning indicative of a successful lesion were split into 2 groups that were balanced with respect to motor response to L-DOPA during screening. One group (controls) received a daily subcutaneous injection of vehicle (0.9% saline containing 0.1 mg/kg ascorbate; 1 ml/kg) for 14 days and the other group (8-OHDPAT) received a daily subcutaneous injection of R(+)-8-OHDPAT (0.3 mg/kg in 1 ml/kg volume) for 14 days. Beginning on day 15, both groups of rats were tested for motor activity and dyskinetic activity in response to the D1 receptor partial agonist SKF 38393 or the D2 receptor agonist quinpirole. Such testing was carried out in a randomized fashion with different doses of dopamine agonist being tested every 4th day (e.g. 3 days between dopamine agonist administration). Throughout this testing phase the control group continued to receive daily injection of vehicle and the 8-OHDPAT group continued to receive daily injection of 0.3 mg/kg R(+)-8-OHDPAT. On days when dopamine agonists were tested the vehicle or R(+)-8-OHDPAT was administered simultaneously with the dopamine agonist.
  • Results. FIGS. 2 and 3 illustrate that administration of a 5-HT1A agonist enhances the anti-parkinsonian action of D1 and D2 agonists, as determined by quantitating contralateral turning in the hemi-lesioned 6-OHDA-treated rat. In both experiments animals were given the 5-HT1A agonist R(+)-8-OHDPAT (0.3 mg/kg) or vehicle (0.9% saline containing ascorbate) for 14 days. Rats that received R(+)-8-OHDPAT for 14 days were then given R(+)-8-OHDPAT (0.3 mg/kg) plus a dopamine agonist on test day, and were observed for 3 hr for anti-parkinsonian effect (contralateral turning). Rats that received vehicle for 14 days were then given a vehicle plus the dopamine agonist on test day, and were observed for 3 hr for anti-parkinsonian effect (contralateral turning). FIG. 2 depicts the data obtained when the D1 agonist SKF 38393 (0.1 mg/kg) was administered on test day. Animals that received R(+)-8-OHDPAT exhibited pronounced contralateral turning when SKF 38393 was administered. This turning behavior was significantly greater than what was observed for animals that received vehicle (p<0.005). In fact, animals that received vehicle plus SKF 38393 displayed no contralateral turning. FIG. 3 depicts the data obtained when the D2 agonist quinpirole (0.05 mg/kg) was administered on test day. Animals that received R(+)-8-OHDPAT exhibited significantly greater contralateral turning following quinpirole as compared to animals that received vehicle plus quinpirole (p<0.001).
  • EXAMPLE 3 The Anti-Parkinsonian Effect of a Serotonin Agonist Requires Repeated Administration
  • Repeated Administration of a 5-HT1A Agonist. The methods described in the previous examples were used to examine the effects of acute and chronic administration of the 5-HT1A agonist, R(+)-8-OHDPAT. The dose of both R(+)-8-OHDPAT and SKF 38393 was 0.3 mg/kg throughout this experiment. FIG. 4 graphically depicts the results of an experiment in which animals received either a) 14 days of R(+)-8-OHDPAT followed by SKF 38393 plus vehicle on test day, b) 14 days of R(+)-8-OHDPAT followed by SKF 38393 plus R(+)-8-OHDPAT on test day, c) 14 days of vehicle followed by SKF 38393 plus vehicle on test day, or d) 14 days of vehicle followed by SKF 38393 plus R(+)-8-OHDPAT on test day. Animals that received 14 day pretreatment with R(+)-8-OHDPAT plus concomitant R(+)-8-OHDPAT +SKF 38393 on test day exhibited significant anti-parkinsonian action greater than all other treatment groups (p<0.001). These data indicate that brain adaptations arising as a result of chronic treatment with R(+)-8-OHDPAT are necessary, but not sufficient, to enhance the anti-parkinsonian actions of dopamine agonists. These data also indicate that concomitant administration of a 5-HT1A agonist plus a dopamine agonist is necessary, but not sufficient, to enhance the anti-parkinsonian actions of dopamine agonists. Thus as determined during development of the present invention, both chronic and concomitant administration of a 5-HT1A agonist are required to achieve enhanced anti-parkinsonian actions.
  • Administration of a 5-HT1A Agonist Synergizes With a Low Dose DA Agonist. Animals were unilaterally lesioned with 6-OHDA and allowed to recover and prescreened with L-DOPA (to assess extent of lesion) as described previously. FIG. 10 depicts the rotational response on day 15 in rats given once daily administration of vehicle+SKF 38393 (0.1, 0.3 or 1.0 mg/kg). There is little net contralateral turning by day 15 of treatment. In contrast, animals receiving once daily R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 treatments displayed a significantly greater contralateral turning response that displayed a dose-dependency with respect to the dose of SKF administered. These data clearly show the ability of a 5-HT1A agonist to synergize with an ineffective dose of a D1 agonist to elicit an anti-parkinsonian response.
  • Previous Exposure to a DA agonist or L-DOPA Is Not Required. Animals were unilaterally lesioned with 6-OHDA and allowed to recover as described previously. However, these animals were not pre-screened with L-DOPA prior to initiation of drug treatments. FIG. 11 compares the rotational response between pre-screened (“primed”) and non-pre-screened (“unprimed”) rats after 15 days of once daily administration of R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 (0.3 mg/kg). There is no significant difference between the groups. These data indicate that the methods and compositions of the present invention comprising combined 5-HT1A agonist and D1 agonist therapy will be effective in Parkinson's disease patients regardless of whether they have previously received treatment (e.g., DA agonist, L-DOPA or nothing).
  • EXAMPLE 4 A Serotonin Agonist Reduces Dyskinetic Behavior Elicited by Moderate Doses of Dopamine Agonists
  • The experiments described in this example were conducted as described above. A dyskinesia index for each animal was determined during a 3 hr rotational test. FIG. 5 illustrates that repeated R(+)-8-OHDPAT administration reduced dyskinetic behavior elicited by a moderately high dose of SKF 38393 (3.0 mg/kg). Similarly, FIG. 6 illustrates that repeated R(+)-8-OHDPAT administration reduced dyskinetic behavior elicited by quinpirole (0.05 mg/kg).
  • EXAMPLE 5 The Anti-Parkinsonian-Enhancing Effect Of R-(+)-8-OHDPAT is Mediated by the 5-HT1A Receptor
  • The methods described in the previous examples were used to examine the effects of a selective 5-HT1A antagonist (WAY 100635) on the enhanced anti-parkinsonian action of a treatment regimen employing both R(+)-8-OHDPAT and SKF 38393. All animals received at least 14 days of daily R(+)-8-OHDPAT. FIG. 7 illustrates that pretreatment of animals on test day with WAY 100635 (0.1 mg/kg) prior to receiving R(+)-8-OHDPAT (0.3 mg/kg) plus SKF 38393 (0.3 mg/kg) blocked the facilitating effects of R(+)-8-OHDPAT on the anti-parkinsonian action of SKF 38393. These data confirm that the facilitating action of R(+)-8-OHDPAT is 5-HT1A receptor-mediated. Nonetheless, knowledge of the mechanism(s) is not necessary in order to make and use the present invention.
  • EXAMPLE 6 Effects of Drug Treatment on Striatal Signaling
  • Transcription factors play a major role in mediating the long-term effects of synaptic plasticity by altering the expression of synapse-associated proteins. ΔFosB is a truncated splice variant of FosB that represents a long-lived transcription factor. ΔFosB has been shown to be upregulated following DA denervation in rodent and primate models of PD, as well as in PD patients (Cenci et al., Neuroscience, 94:515-527, 1999; Perez-Otano et al., Brain Res Mol Brain Res, 53:41-52 1998; and Tekumalla et al., Biol Psychiatry, 50:813-816, 2001). Repeated administration of L-DOPA or a D1 agonist further elevates striatal ΔFosB, while D2 agonists have no effect (Cenci et al., supra, 1999; Andersson et al., Neurobiol Dis, 6:461-474, 1999; and Doucet et al., Eur J Neurosci, 8:365-385, 1996).
  • To test whether a drug-related rise in ΔFosB contributes to dyskinesias elicited by repeated L-DOPA treatment, ΔFosB levels were assessed by immunoblotting. FIG. 8 provides an image of a typical blot. FIG. 9 illustrates that once daily administration of L-DOPA (25 mg/kg plus 6.25 mg/kg benserazide) or SKF 38393 (1 mg/kg) for 15 days elicits a significant increase in ΔFosB in the denervated striatum of unilateral 6-OHDA-lesioned rats (*, significantly greater than vehicle-treated; p<0.05). This figure also demonstrates that concomitant administration of the 5-HT1A agonist R(+)-8-OHDPAT (0.3 mg/kg) with SKF 38393 prevents the D1 agonist-induced rise in ΔFosB (#, significantly less than VEH+SKF; p<0.05 and not significantly different from vehicle-treated). Thus a significant target of the treatments of the present invention is contemplated to be ΔFosB in the striatum. Even so, knowledge of the mechanism(s) is not necessary in order to make and use the present invention.
  • EXAMPLE 7 A Serotonin Agonist Enhances the Anti-Parkinsonian Actions of Dopamine Agonists in a Marmoset Model of Parkinson's Disease
  • This example describes testing a serotonin agonist with a dopamine agonist in a nonhuman primate model of Parkinson's disease. Briefly, adult common marmosets (Callithrix jacchus) received either 0.9% saline SC (1 ml/kg) daily for five consecutive days (naïve controls) or MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride) SC (2 mg/kg) daily for five consecutive days. MPTP administration induces a motor disorder characterized by akinesia, bradykinesia and rigidity, which is relatively stable after an initial recovery period. Abnormal posture, loss of vocalization, diminished blinking, incoordination and a tremor are also frequently observed.
  • Efficacy of administration of a serotonin type 1A receptor agonist and a dopamine receptor agonist in reducing at least one symptom of MPTP-induced motor disorder is measured by behavioral assessment as described (Pearce et al., Psychopharmacol, 142:51-60, 1999). Briefly, automated locomotor counts, behavioral observation and dyskinesia scoring are recorded. Automated locomotor counts are taken through the use of cages equipped with multiple infrared photocells. The number of light beam interruptions caused by movement is enumerated in 10-min intervals over a 2-12 h and recorded by computer. An initial period of 30-60 min is used for habituation to the cages prior to drug dosing and behavioral assessment. The animals are also closely monitored by trained observers (e.g., a clinical neurologist with experience in movement disorders). The degree of motor dysfunction is scored on a disability grading system; alertness (normal/0, abnormal/1); reaction to stimuli (normal/0, reduced/1, slow/2, absent/3); blinking (normal/0, abnormal/1); checking movements (present/0, reduced/1, absent/2); posture (normal/0, abnormal trunk/+1, tail/+1, limbs/+1, grossly abnormal/4); balance (normal/0, unstable/1, spontaneous falls/2); motility (normal/0, mild slowing/1, moderate bradykinesia/2, akinesia/3); vocalisation (normal/0, reduced/1, absent/2); tremor (absent/0, present/1) fur condition (normal/0, dirty/1). These values are summed to yield a disability score out of 20. Disability scores are generated from continuous observation at half hour intervals before and during peak drug effect.
  • Abnormal movements are described according to classically defined criteria: chorea (rapid random kicking limb movements); athetosis (sinuous writhing limb movements); dystonia (abnormal sustained posturing); and stereotypy(repetitive purposeless or semipurposive movement). The presence of dyskinesias is scored at maximum drug effect employing a dyskinesia scoring system: 0=absent; 1=mild (fleeting and rare dyskinetic postures and movements); 2=moderate (more prominent abnormal movements, but not interfering significantly with normal behavior); 3=marked (frequent and at times continuous dyskinesias intruding upon normal repertory of activity); and 4=severe (virtually continuous dyskinetic activity, impairing the animal's normal behavior).
  • The inventors contemplate that administration of a serotonin receptor type 1A receptor agonist with a low-dose dopamine receptor agonist reduces motor dysfunction (reduces parkinsonian symptoms and/or dopamine-replacement therapy-induced dyskinesias) in MPTP-lesioned marmosets.
  • EXAMPLE 8 A Serotonin Agonist Enhances the Anti-Parkinsonian Actions of Dopamine Agonists in a Parkinson's Disease Patients
  • This example describes testing a serotonin agonist with a dopamine agonist in a human Parkinson's disease patients. Briefly, all patients admitted to this study are diagnosed with idiopathic Parkinson's disease (See, e.g., Gibb and Lees, J Neurol Neurosurg Psychiatry, 51:745-752, 1988).
  • Efficacy of administration of a serotonin type 1A receptor agonist and a dopamine receptor agonist in reducing at least one symptom of Parkinson's disease is measured using the United Parkinson's Disease Rating Scale (Fahn et al. (eds.), Recent Developments in Parkinson's Disease, vol. 2, New York, N.Y.: Macmillan Publishing Co. Inc., pp. 153-163 and 293-304, 1987), whereas dyskinesias are assessed using an art recognized severity scale (Marconi et al., Mov Disord, 9:2-12, 1994). Briefly, the dyskinesia severity scale rates abnormal movements from 0 (none) to 4 (severe with markedly impaired function) in six different parts of the body (face, neck and trunk, and four limbs).
  • The inventors contemplate that administration of a serotonin receptor type 1A receptor agonist with a low-dose dopamine receptor agonist reduces motor dysfunction (reduces parkinsonian symptoms and/or dopamine-replacement therapy-induced dyskinesias) in human Parkinson's disease patients.
  • All publications and patents mentioned in the above specification are herein incorporated by reference. Various modifications and variations of the described method and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention, which are obvious to those skilled in the relevant fields, are intended to be within the scope of the following claims.

Claims (25)

1. A method of treating a motor disorder, comprising:
a) providing:
i) a serotonin type 1A (5-HT1A) receptor agonist,
ii) a dopamine receptor agonist, and
iii) a subject diagnosed as having a motor disorder; and
b) administering said serotonin type 1A receptor (5-HT1A) agonist to said subject during a pre-conditioning period; and
c) co-administering said serotonin type 1A receptor (5-HT1A) agonist in combination with a low dose of said dopamine receptor agonist to said subject, under conditions suitable for reducing at least one symptom of said motor disorder, wherein said low dose of said dopamine receptor agonist is below that required for reducing at least one symptom of said motor disorder when administered in the absence of said serotonin type 1A (5-HT1A) receptor agonist.
2. The method of claim 1, wherein said motor disorder is Parkinson's disease.
3. The method of claim 1, wherein said motor disorder is parkinsonism arising from viral infection or drug use.
4. The method of claim 2, wherein said dopamine receptor agonist is not a dopamine precursor.
5. The method of claim 2, wherein said serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of arylpiperazines, azapirones, aminotetralins, and aminochromanes.
6. The method of claim 2, wherein said serotonin type 1A (5-HT1A) receptor agonist is 8-hydroxy-2-(di-n-propylamino)tetralin (8OH-DPAT).
7. The method of claim 2, wherein said serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of tandospirone, ipsapirone, gepirone, flibanserin, flesinoxan, sarizotan, repinotan and PRX-00023.
8. The method of claim 2, wherein said dopamine receptor agonist is a D1 type dopamine receptor agonist.
9. The method of claim 8, wherein said D1 type dopamine receptor agonist is selected from the group consisting of SKF38393, SKF83959, SKF81297, SKF77434, SKF75670, SKF82958, and derivatives thereof.
10. The method of claim 8, wherein said D1 type dopamine receptor agonist is selected from the group consisting of dihydrexidine, dinapsoline, A-77636, ABT-431, CY208-243, and A-68930.
11. The method of claim 2, wherein said dopamine receptor agonist is a D2 type dopamine receptor agonist.
12. The method of claim 11, wherein said a D2 type dopamine receptor agonist is quinpirole.
13. The method of claim 11, wherein said a D2 type dopamine receptor agonist is selected from the group consisting of quinelorane, terguride, pergolide, talipexole, pramipexole, sumanirole, ropinirole, roxindole, bromocriptine, and cabergoline.
14. The method of claim 2, wherein said dopamine agonist is a dopamine precursor.
15. The method of claim 14, wherein said dopamine precursor is levodopa (L-DOPA), and wherein said L-DOPA is administered with one or both of a L-aromatic amino acid decarboxylase inhibitor and a catechol-O-methyltransferase (COMT) inhibitor.
16. The method of claim 2, wherein one or both of said serotonin type 1A (5-HT1A) receptor agonist and said dopamine receptor agonist is a D1 type dopamine receptor agonist.
17. The method of claim 16, wherein said serotonin type 1A (5-HT1A) receptor agonist is selected from the group consisting of sunipetron and aripiprazole.
18. The method of claim 2, wherein step b further comprises administering a low dose of said dopamine receptor agonist to said subject during said pre-conditioning period, wherein said low dose is of said dopamine receptor agonist is below that required for reducing at least one symptom of said motor disorder when administered in the absence of said serotonin type 1A (5-HT1A) receptor agonist.
19. The method of claim 2, wherein said at least one symptom comprises bradykinesia.
20. The method of claim 2, wherein said at least one symptom comprises tremor.
21. The method of claim 2, wherein said at least one symptom comprises muscle rigidity.
22. The method of claim 2, wherein said co-administering said serotonin type 1A receptor (5-HT1A) agonist in combination with said dopamine receptor agonist to said subject is under conditions further suitable for reducing at least one side effect of said dopamine receptor agonist.
23. The method of claim 22, wherein said at least one side effect of said dopamine receptor agonist comprises dyskinesia.
24. A method of treating a Parkinson's disease, comprising:
a) providing:
i) a serotonin type 1A (5-HT1A) receptor agonist,
ii) a dopamine receptor agonist, wherein said dopamine receptor agonist is not a dopamine precursor, and
iii) a subject diagnosed as having Parkinson's disease; and
b) administering said serotonin type 1A receptor (5-HT1A) agonist to said subject during a pre-conditioning period; and
c) co-administering said serotonin type 1A receptor (5-HT1A) agonist in combination with a low dose of said dopamine receptor agonist to said subject, under conditions suitable for reducing at least one symptom of said Parkinson's disease, wherein said low dose is of said dopamine receptor agonist is below that required for reducing at least one symptom of said Parkinson's disease when administered in the absence of said serotonin type 1A (5-HT1A) receptor agonist.
25. A method for treating a subject, comprising:
a) providing;
i) a serotonin type 1A (5-HT1A) receptor agonist,
ii) a dopamine receptor agonist,
iii) a subject receiving said dopamine receptor agonist for treatment of Parkinson's disease, wherein said subject is suffering from dopamine receptor agonist-induced dyskinesia;
b) co-administering said serotonin type 1A receptor (5-HT1A) agonist in combination with said dopamine receptor agonist to said subject during a preconditioning period under conditions suitable for reducing said dopamine receptor agonist-induced dyskinesia;
c) co-administering said serotonin type 1A receptor (5-HT1A) agonist in combination with said dopamine receptor agonist to said subject during a dosage optimization period, wherein dosage of said dopamine receptor agonist is gradually reduced until at least one symptom of said Parkinson's disease is re-emerges; and
d) co-administering said serotonin type 1A receptor (5-HT1A) agonist in combination with said dopamine receptor agonist to said subject under conditions suitable for reducing said at least one symptom of said Parkinson's disease, while reducing said dopamine receptor agonist-induced dyskinesia, wherein dosage of said dopamine receptor agonist is below that of step b and above that of step c.
US11/713,156 2006-02-28 2007-02-28 Pharmacological treatment of Parkinson's disease Active 2029-04-25 US9066903B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US11/713,156 US9066903B2 (en) 2006-02-28 2007-02-28 Pharmacological treatment of Parkinson's disease
US12/730,972 US9226904B2 (en) 2006-02-28 2010-03-24 Pharmacological treatment of Parkinson's disease
US14/958,430 US20160082006A1 (en) 2006-02-28 2015-12-03 Pharmacological treatment of parkinson's disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US77793906P 2006-02-28 2006-02-28
US11/713,156 US9066903B2 (en) 2006-02-28 2007-02-28 Pharmacological treatment of Parkinson's disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/730,972 Continuation US9226904B2 (en) 2006-02-28 2010-03-24 Pharmacological treatment of Parkinson's disease

Publications (2)

Publication Number Publication Date
US20070249621A1 true US20070249621A1 (en) 2007-10-25
US9066903B2 US9066903B2 (en) 2015-06-30

Family

ID=38620235

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/713,156 Active 2029-04-25 US9066903B2 (en) 2006-02-28 2007-02-28 Pharmacological treatment of Parkinson's disease
US12/730,972 Active 2028-04-25 US9226904B2 (en) 2006-02-28 2010-03-24 Pharmacological treatment of Parkinson's disease
US14/958,430 Abandoned US20160082006A1 (en) 2006-02-28 2015-12-03 Pharmacological treatment of parkinson's disease

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/730,972 Active 2028-04-25 US9226904B2 (en) 2006-02-28 2010-03-24 Pharmacological treatment of Parkinson's disease
US14/958,430 Abandoned US20160082006A1 (en) 2006-02-28 2015-12-03 Pharmacological treatment of parkinson's disease

Country Status (1)

Country Link
US (3) US9066903B2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009151625A1 (en) * 2008-06-13 2009-12-17 Teva Pharmaceutical Industries, Ltd. Rasagiline for parkinson's disease modification
WO2009156380A1 (en) * 2008-06-24 2009-12-30 Bjoerklund Anders Eltoprazine for suppression of l-dopa induced dyskinesias
JP2011521954A (en) * 2008-05-30 2011-07-28 サイコジェニックス・インコーポレーテッド Treatment of nerve and psychiatric disorders
EP2373314A1 (en) * 2008-12-05 2011-10-12 Merz Pharma GmbH & Co. KGaA Use of eltoprazine for the treatment of l-dopa-induced dyskinesia
WO2012048710A1 (en) 2010-10-15 2012-04-19 Concit Pharma Aps Combinations of serotonin receptor agonists for treatment of movement disorders
WO2012163365A1 (en) 2011-06-01 2012-12-06 Concit Pharma Aps Combinations of serotonin receptor agonists for treatment of movement disorders
US20130017274A1 (en) * 2011-03-16 2013-01-17 Buck Institute For Research On Aging Low dose lithium in the treatment or prophylaxis of parkinson's disease
US10561618B2 (en) 2012-04-18 2020-02-18 Contera Pharma Aps Orally available pharmaceutical formulation suitable for improved management of movement disorders
US11548893B2 (en) 2017-07-15 2023-01-10 Arisan Therapeutics Inc. Enantiomerically pure adamantane carboxamides for the treatment of filovirus infection

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112013014304A2 (en) * 2010-12-10 2016-07-19 Synagile Corp subcutaneously infusible levodopa prodrug compositions and method of infusion
CA2973701A1 (en) * 2015-01-13 2016-07-21 Amarantus Bioscience Holdings, Inc. Methods of treating motor and movement disorders and side effects thereof associated with parkinson's disease treatments
AU2016258179B2 (en) 2015-05-06 2021-07-01 Synagile Corporation Pharmaceutical suspensions containing drug particles, devices for their administration, and methods of their use

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5712270A (en) * 1995-11-06 1998-01-27 American Home Products Corporation 2-arylamidothiazole derivatives with CNS activity
US6300329B1 (en) * 1998-09-21 2001-10-09 Mclean Stafford Pharmaceutical agents for the treatment of Parkinson's disease, ADHD and microadenomas
US20020177616A1 (en) * 2001-01-16 2002-11-28 Thomas Fahrig Use of chromans
US6518272B2 (en) * 1998-06-15 2003-02-11 Wyeth Cycloalkyl-substituted aryl-piperazines, piperidines and tetrahydropyridines as serotonergic agents
US6521623B1 (en) * 2000-09-19 2003-02-18 Boehringer Ingelheim Pharma Kg N,N'-disubstituted benzimidazolone derivatives with affinity at the serotonin and dopamine receptors
US20040067956A1 (en) * 2000-11-03 2004-04-08 Jonathan Brotchie Treatment of motor fluctuations
US20040198753A1 (en) * 2002-01-28 2004-10-07 Hiroshi Kase Methods of treating patients suffering from movement disorders
US6916823B2 (en) * 2001-01-16 2005-07-12 Purdue Research Foundation Method of treatment of dopamine-related dysfunction
US20050239797A1 (en) * 1999-11-05 2005-10-27 Smithkline Beecham, Plc Isoquinoline and quinazoline derivatives having a combined 5HT1A, 5HT1B, and 5HT1D receptor activity
US20090156581A1 (en) * 2005-04-15 2009-06-18 Board Of Trustrees Of Michigan State University Aminergic pharmaceutical compositions and methods

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU577802B2 (en) 1983-10-17 1988-10-06 Duphar International Research B.V. Blood-pressure lowering piperazine derivatives
DE3586794T2 (en) 1984-12-21 1993-05-27 Duphar Int Res MEDICINAL PRODUCTS WITH PSYCHOTROPER EFFECT.
EP0190472B1 (en) 1984-12-21 1989-07-12 Duphar International Research B.V New pharmaceutical compositions having anti-psychotic properties
DE3782525T2 (en) 1986-02-27 1993-05-27 Duphar Int Res ARYL-SUBSTITUTED (N-PIPERIDINYL) METHYL AND (N-PIPERAZINYL) METHYLAZOLES WITH ANTIPSYCHOTIC EFFECT.
DK172628B1 (en) 1987-09-11 1999-08-02 Duphar Int Res Use of piperazine derivatives for the manufacture of drugs for the treatment of fear or fear syndromes
GB9125900D0 (en) 1991-12-05 1992-02-05 Wyeth John & Brother Ltd Piperazine derivatives
AU3241093A (en) 1992-01-07 1993-08-03 Merrell Dow Pharmaceuticals Inc. Treatment of involuntary movements with 5HT-1A receptor agonists
RU2118322C1 (en) 1993-07-05 1998-08-27 Дюфар Интернэшнл Рисерч Б.В. 2,3-dihydro-1,4-benzodioxine-5-yl-pyrerazine derivatives and salts thereof
JPH11511156A (en) 1995-08-11 1999-09-28 スミスクライン・ビーチャム・パブリック・リミテッド・カンパニー Biphenyl (thio) amide and biphenylethane (thio) one derivatives, their preparation and their use as 5-HT 1D receptor antagonists
GB9706089D0 (en) 1997-03-24 1997-05-14 Scherer Ltd R P Pharmaceutical composition
DE19730989A1 (en) 1997-07-18 1999-01-21 Merck Patent Gmbh Piperazine derivatives
SE9703378D0 (en) 1997-09-18 1997-09-18 Astra Ab New compounds
US6967201B1 (en) 1999-07-29 2005-11-22 Eli Lilly And Company Benzofurylpiperazines and benzofurylhomopiperazines: serotonin agonists
KR100634039B1 (en) 1999-08-23 2006-10-17 솔베이 파마슈티칼스 비. 브이 New phenylpiperazines
UA73981C2 (en) 2000-03-10 2005-10-17 Merck Patent Gmbh (r)-(-)-2-[5-(4-fluorophenyl)-3-pyridylmethylaminomethyl]-chromane for treatment of extrapyramidal movement disorders (variants), pharmaceutical composition and kit
EP1284731A1 (en) 2000-05-12 2003-02-26 Solvay Pharmaceuticals B.V. Use of compounds having combined dopamine d2, 5-ht1a and alpha adrenoreceptor agonistic action for treating cns disorders
AR032712A1 (en) 2001-02-21 2003-11-19 Solvay Pharm Bv A MESILATE OF PHENYLPIPERAZINE DERIVATIVES AND PHARMACEUTICAL COMPOSITIONS CONTAINING IT
EP1256343B1 (en) 2001-05-11 2006-07-05 Jürgen K. Dr. Beck Flibanserin for the treatment of extrapyramidal movement disorders
JP5080716B2 (en) 2001-07-20 2012-11-21 サイコジェニックス・インコーポレーテッド Treatment of attention deficit / hyperactivity disorder
US20040147581A1 (en) 2002-11-18 2004-07-29 Pharmacia Corporation Method of using a Cox-2 inhibitor and a 5-HT1A receptor modulator as a combination therapy
US20070179168A1 (en) 2005-11-28 2007-08-02 Orexigen Therapeutics, Inc. Methods of treating anxiety disorders
US20110183995A1 (en) * 2008-06-24 2011-07-28 Neurosearch A/S Eltoprazine for suppression of l-dopa induced dyskinesias

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5712270A (en) * 1995-11-06 1998-01-27 American Home Products Corporation 2-arylamidothiazole derivatives with CNS activity
US6518272B2 (en) * 1998-06-15 2003-02-11 Wyeth Cycloalkyl-substituted aryl-piperazines, piperidines and tetrahydropyridines as serotonergic agents
US6300329B1 (en) * 1998-09-21 2001-10-09 Mclean Stafford Pharmaceutical agents for the treatment of Parkinson's disease, ADHD and microadenomas
US6608064B2 (en) * 1998-09-21 2003-08-19 Pfizer Inc Pharmaceutical agents for the treatment of Parkinson's disease, ADHD and microadenomas
US20050239797A1 (en) * 1999-11-05 2005-10-27 Smithkline Beecham, Plc Isoquinoline and quinazoline derivatives having a combined 5HT1A, 5HT1B, and 5HT1D receptor activity
US6521623B1 (en) * 2000-09-19 2003-02-18 Boehringer Ingelheim Pharma Kg N,N'-disubstituted benzimidazolone derivatives with affinity at the serotonin and dopamine receptors
US20040067956A1 (en) * 2000-11-03 2004-04-08 Jonathan Brotchie Treatment of motor fluctuations
US20110178178A1 (en) * 2000-11-03 2011-07-21 Motac Neuroscience Limited Treatment of Motor Fluctuations
US20020177616A1 (en) * 2001-01-16 2002-11-28 Thomas Fahrig Use of chromans
US6916823B2 (en) * 2001-01-16 2005-07-12 Purdue Research Foundation Method of treatment of dopamine-related dysfunction
US20040198753A1 (en) * 2002-01-28 2004-10-07 Hiroshi Kase Methods of treating patients suffering from movement disorders
US20090156581A1 (en) * 2005-04-15 2009-06-18 Board Of Trustrees Of Michigan State University Aminergic pharmaceutical compositions and methods

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2374459A1 (en) * 2008-05-30 2011-10-12 Psychogenics Inc. Treatment for neurological and mental disorders
KR101746756B1 (en) * 2008-05-30 2017-06-27 싸이코제닉스 아이엔씨. Treatment for neurological and mental disorders
JP2011521954A (en) * 2008-05-30 2011-07-28 サイコジェニックス・インコーポレーテッド Treatment of nerve and psychiatric disorders
JP2015155439A (en) * 2008-05-30 2015-08-27 サイコジェニックス・インコーポレーテッドPsychogenics Inc. Treatment for neurological and mental disorders
US20090312436A1 (en) * 2008-06-13 2009-12-17 Ruth Levy Rasagiline for parkinson's disease modification
WO2009151625A1 (en) * 2008-06-13 2009-12-17 Teva Pharmaceutical Industries, Ltd. Rasagiline for parkinson's disease modification
US20110183995A1 (en) * 2008-06-24 2011-07-28 Neurosearch A/S Eltoprazine for suppression of l-dopa induced dyskinesias
WO2009156380A1 (en) * 2008-06-24 2009-12-30 Bjoerklund Anders Eltoprazine for suppression of l-dopa induced dyskinesias
EP2373314A1 (en) * 2008-12-05 2011-10-12 Merz Pharma GmbH & Co. KGaA Use of eltoprazine for the treatment of l-dopa-induced dyskinesia
WO2012048710A1 (en) 2010-10-15 2012-04-19 Concit Pharma Aps Combinations of serotonin receptor agonists for treatment of movement disorders
US9186359B2 (en) 2010-10-15 2015-11-17 Contera Pharma Aps Combinations of serotonin receptor agonists for treatment of movement disorders
US10632116B2 (en) 2010-10-15 2020-04-28 Contera Pharma A/S Combinations of serotonin receptor agonists for treatment of movement disorders
US20130017274A1 (en) * 2011-03-16 2013-01-17 Buck Institute For Research On Aging Low dose lithium in the treatment or prophylaxis of parkinson's disease
WO2012163365A1 (en) 2011-06-01 2012-12-06 Concit Pharma Aps Combinations of serotonin receptor agonists for treatment of movement disorders
US10561618B2 (en) 2012-04-18 2020-02-18 Contera Pharma Aps Orally available pharmaceutical formulation suitable for improved management of movement disorders
US11548893B2 (en) 2017-07-15 2023-01-10 Arisan Therapeutics Inc. Enantiomerically pure adamantane carboxamides for the treatment of filovirus infection

Also Published As

Publication number Publication date
US20100179171A1 (en) 2010-07-15
US9226904B2 (en) 2016-01-05
US9066903B2 (en) 2015-06-30
US20160082006A1 (en) 2016-03-24

Similar Documents

Publication Publication Date Title
US9066903B2 (en) Pharmacological treatment of Parkinson&#39;s disease
Tahar et al. Effect of a selective glutamate antagonist on L-dopa-induced dyskinesias in drug-naive parkinsonian monkeys
US20180221377A1 (en) Methods of treating patients suffering from movement disorders
Jankovic et al. Current approaches to the treatment of Parkinson’s disease
Levandis et al. Systemic administration of an mGluR5 antagonist, but not unilateral subthalamic lesion, counteracts l-DOPA-induced dyskinesias in a rodent model of Parkinson's disease
Rasmussen et al. A selective AMPA antagonist, LY293558, suppresses morphine withdrawal-induced activation of locus coeruleus neurons and behavioral signs of morphine withdrawal
US6232326B1 (en) Treatment for schizophrenia and other dopamine system dysfunctions
US6911475B1 (en) Use of nicotine or its derivatives in a drug for treating neurological disease, in particular Parkinson&#39;s disease
JP2014074032A (en) Mglu receptor antagonist for treating disorder associated with mglu receptor including addiction and depression
Errico et al. Higher free D-aspartate and N-methyl-D-aspartate levels prevent striatal depotentiation and anticipate L-DOPA-induced dyskinesia
AU2003207734A1 (en) Methods of treating patients suffering from movement disorders
Stocchi Continuous dopaminergic stimulation and novel formulations of dopamine agonists
JP2009137995A (en) Substituted aminoalkanephosphonic acid for treating neuropathic pain, affective and attention disorder, schizophrenia, tinnitus, myopia and other ocular disorder
WO2018013951A1 (en) Targeting adenosine a2a receptors for the treatment of levodopa-induced dyskinesias
Metman et al. Role of surgery in the treatment of motor complications
Fisher et al. Differential effects of NMDA and non-NMDA antagonists on the activity of aromatic L-amino acid decarboxylase activity in the nigrostriatal dopamine pathway of the rat
EP2037964A2 (en) Combination preparations comprising bifeprunox and l-dopa
Fornal et al. Pindolol, a Putative 5-Hydroxytryptamine1AAntagonist, Does Not Reverse the Inhibition of Serotonergic Neuronal Activity Induced by Fluoxetine in Awake Cats: Comparison to WAY-100635
Jonkers et al. MK801 influences L-DOPA-induced dopamine release in intact and hemi-parkinson rats
US7915262B2 (en) Combination preparations comprising SLV308 and a dopamine agonist
Stocchi Dopamine receptor agonists in the treatment of advanced Parkinson's disease
Nomoto et al. Inter-and intra-individual variation in L-dopa pharmacokinetics in the treatment of Parkinson's disease
EP2035002A1 (en) Combination preparations comprising slv308 and a l-dopa
Kase et al. Administering adenosine A2A receptor antagonist to reduce or suppress side effects of Parkinson’s disease therapy
Jenner Novel Therapeutic Approaches to the Treatment of Parkinson's Disease

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNITED STATES GOVERNMENT AS REPRESENTED BY THE DEP

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WOLF, WILLIAM A.;ISTRE, ERIK D.;REEL/FRAME:019505/0155;SIGNING DATES FROM 20070608 TO 20070610

Owner name: UNITED STATES GOVERNMENT AS REPRESENTED BY THE DEP

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WOLF, WILLIAM A.;ISTRE, ERIK D.;SIGNING DATES FROM 20070608 TO 20070610;REEL/FRAME:019505/0155

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: SURCHARGE FOR LATE PAYMENT, LARGE ENTITY (ORIGINAL EVENT CODE: M1554); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8